101
|
Cheng LF, You CQ, Peng C, Ren JJ, Guo K, Liu TL. Mesenchymal stem cell-derived exosomes as a new drug carrier for the treatment of spinal cord injury: A review. Chin J Traumatol 2024; 27:134-146. [PMID: 38570272 PMCID: PMC11138942 DOI: 10.1016/j.cjtee.2024.03.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 02/20/2024] [Accepted: 03/15/2024] [Indexed: 04/05/2024] Open
Abstract
Spinal cord injury (SCI) is a devastating traumatic disease seriously impairing the quality of life in patients. Expectations to allow the hopeless central nervous system to repair itself after injury are unfeasible. Developing new approaches to regenerate the central nervous system is still the priority. Exosomes derived from mesenchymal stem cells (MSC-Exo) have been proven to robustly quench the inflammatory response or oxidative stress and curb neuronal apoptosis and autophagy following SCI, which are the key processes to rescue damaged spinal cord neurons and restore their functions. Nonetheless, MSC-Exo in SCI received scant attention. In this review, we reviewed our previous work and other studies to summarize the roles of MSC-Exo in SCI and its underlying mechanisms. Furthermore, we also focus on the application of exosomes as drug carrier in SCI. In particular, it combs the advantages of exosomes as a drug carrier for SCI, imaging advantages, drug types, loading methods, etc., which provides the latest progress for exosomes in the treatment of SCI, especially drug carrier.
Collapse
Affiliation(s)
- Lin-Fei Cheng
- Medical College, Anhui University of Science and Technology, Huainan, 232000, Anhui province, China
| | - Chao-Qun You
- Department of Orthopaedic Oncology, Changzheng Hospital, Navy Medical University, Shanghai, 200003, China
| | - Cheng Peng
- Department of Orthopaedic Oncology, Changzheng Hospital, Navy Medical University, Shanghai, 200003, China
| | - Jia-Ji Ren
- Department of Orthopaedic Oncology, Changzheng Hospital, Navy Medical University, Shanghai, 200003, China
| | - Kai Guo
- Department of Orthopaedics, The Central Hospital of Shanghai Putuo District, Shanghai, 200333, China
| | - Tie-Long Liu
- Medical College, Anhui University of Science and Technology, Huainan, 232000, Anhui province, China.
| |
Collapse
|
102
|
Zhang J, Li Z, Chen Z, Shi W, Xu Y, Huang Z, Lin Z, Dou R, Lin S, Jiang X, Li M, Jiang S. Comprehensive analysis of macrophage-related genes in prostate cancer by integrated analysis of single-cell and bulk RNA sequencing. Aging (Albany NY) 2024; 16:6809-6838. [PMID: 38663915 PMCID: PMC11087116 DOI: 10.18632/aging.205727] [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: 10/30/2023] [Accepted: 01/30/2024] [Indexed: 05/08/2024]
Abstract
Macrophages, as essential components of the tumor immune microenvironment (TIME), could promote growth and invasion in many cancers. However, the role of macrophages in tumor microenvironment (TME) and immunotherapy in PCa is largely unexplored at present. Here, we investigated the roles of macrophage-related genes in molecular stratification, prognosis, TME, and immunotherapeutic response in PCa. Public databases provided single-cell RNA sequencing (scRNA-seq) and bulk RNAseq data. Using the Seurat R package, scRNA-seq data was processed and macrophage clusters were identified automatically and manually. Using the CellChat R package, intercellular communication analysis revealed that tumor-associated macrophages (TAMs) interact with other cells in the PCa TME primarily through MIF - (CD74+CXCR4) and MIF - (CD74+CD44) ligand-receptor pairs. We constructed coexpression networks of macrophages using the WGCNA to identify macrophage-related genes. Using the R package ConsensusClusterPlus, unsupervised hierarchical clustering analysis identified two distinct macrophage-associated subtypes, which have significantly different pathway activation status, TIME, and immunotherapeutic efficacy. Next, an 8-gene macrophage-related risk signature (MRS) was established through the LASSO Cox regression analysis with 10-fold cross-validation, and the performance of the MRS was validated in eight external PCa cohorts. The high-risk group had more active immune-related functions, more infiltrating immune cells, higher HLA and immune checkpoint gene expression, higher immune scores, and lower TIDE scores. Finally, the NCF4 gene has been identified as the hub gene in MRS using the "mgeneSim" function.
Collapse
Affiliation(s)
- Jili Zhang
- Department of Urology, The First Navy Hospital of Southern Theater Command, Zhanjiang, Guangdong, China
| | - Zhihao Li
- Center of Reproductive Medicine, Fujian Maternity and Child Health Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Zhenlin Chen
- Department of Urology, Fujian Union Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Wenzhen Shi
- Department of Urology, Fujian Union Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Yue Xu
- Department of Urology, Fujian Union Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Zhangcheng Huang
- Department of Urology, Fujian Union Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Zequn Lin
- Department of Urology, Fujian Union Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Ruiling Dou
- Department of Urology, Fujian Union Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Shaoshan Lin
- Department of Urology, Fujian Union Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Xin Jiang
- Department of Urology, The First Navy Hospital of Southern Theater Command, Zhanjiang, Guangdong, China
| | - Mengqiang Li
- Department of Urology, Fujian Union Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Shaoqin Jiang
- Department of Urology, Fujian Union Hospital, Fujian Medical University, Fuzhou, Fujian, China
| |
Collapse
|
103
|
He Y, Tang Y, Wen S, Dong L, Li F, Deng Y, Tao Z. LINC00998 Modulating M2 Macrophage Activation in Allergic Rhinitis by Stabilizing BOB.1 mRNA. J Inflamm Res 2024; 17:2309-2326. [PMID: 38638161 PMCID: PMC11026101 DOI: 10.2147/jir.s444692] [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: 11/01/2023] [Accepted: 04/09/2024] [Indexed: 04/20/2024] Open
Abstract
Background Allergic rhinitis (AR) is globally recognized as a considerable threat to human health with a rising prevalence and a substantial medical and socioeconomic burden. Numerous studies have emphasized the significance of long noncoding RNAs (lncRNAs) in allergic responses. Hence, this research dealt with exploring the involvement of the lncRNA LINC00998 in the mechanism of AR. Methods LINC00998 expression was assessed by qRT-PCR in peripheral blood mononuclear cells acquired from individuals with AR. Additionally, the potential relationship between LINC00998 and macrophage polarization was observed in vitro. Then we constructed AR mice model and macrophage polarization models using THP-1 cells as well as primary human macrophages to verify the M2 shift in AR and the low expression level of LINC00998 in M2 macrophages. We used gain- and loss-of-function experiments to explore the modification of LINC00998 in macrophage polarization. Furthermore, we explored the underlying mechanism of LINC00998 mediates through qRT-PCR, flow cytometry, and Western blot. Results The analysis revealed a significant decrease in LINC00998 expression in the samples obtained from patients with AR. LINC00998 is markedly increased in M1 macrophages whereas decreased in M2 macrophages in vitro. Furthermore, suppression of LINC00998 caused a remarkable enhancement in M2 polarization, whereas its overexpression led to its attenuation. Knockdown of LINC00998 led to a remarkable downregulation of BOB.1 mRNA and protein, while overexpression of LINC00998 upregulated their expression. Moreover, it was found that BOB.1 modulated macrophage polarization through the PU.1/IL-1β axis. Meanwhile, the modulation of LINC00098 overexpression on macrophage polarization and PU.1/ IL-1β can be reversed by BOB.1 siRNA. Conclusion This research revealed the lncRNA LINC00998 altered M2 macrophage polarization by regulating the BOB.1/PU.1/IL-1β axis, which open up new avenues for studying the pathogenesis of AR.
Collapse
Affiliation(s)
- Yan He
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, People’s Republic of China
| | - Yulei Tang
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, People’s Republic of China
| | - Silu Wen
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, People’s Republic of China
| | - Lin Dong
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, People’s Republic of China
| | - Fen Li
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, People’s Republic of China
- Research Institute of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, People’s Republic of China
| | - Yuqing Deng
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, People’s Republic of China
| | - Zezhang Tao
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, People’s Republic of China
| |
Collapse
|
104
|
Cui Y, Li Z, Ni L, Yu S, Shan X, Hu P, Ji Z, Jing W, Zhou Y, Wang B, Dong H, Zhou J, Xie K, Yu Q. Induction of MTHFD2 in Macrophages Inhibits Reactive Oxygen Species-mediated NF-κB Activation and Protects against Inflammatory Responses. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:1345-1356. [PMID: 38407485 DOI: 10.4049/jimmunol.2300209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 02/06/2024] [Indexed: 02/27/2024]
Abstract
The one-carbon metabolism enzyme methylenetetrahydrofolate dehydrogenase 2 (MTHFD2) is critical for cancer cell proliferation and immune cell phenotypes, but whether it can contribute to macrophage inflammatory responses remains unclear. In this study, we show that MTHFD2 was upregulated by LPS in murine macrophages upon activation of the TLR4-MyD88-IKKα/β-NF-κB signaling pathway. MTHFD2 significantly attenuated LPS-induced macrophage proinflammatory cytokine production through its enzymatic activity. Notably, ablation of myeloid MTHFD2 rendered mice more sensitive to septic shock and CCl4-induced acute hepatitis. Mechanistically, MTHFD2 restrained IKKα/β-NF-κB activation and macrophage inflammatory phenotype by scavenging reactive oxygen species through the generation of NADPH. Our study reveals MTHFD2 as a "self-control" mechanism in macrophage-mediated inflammatory responses.
Collapse
Affiliation(s)
- Yan Cui
- Tianjin Institute of Immunology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, School of Basic Medical Sciences, Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, Tianjin Medical University, Tianjin, China
- Department of Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin, China
- Department of Anesthesiology, Tianjin Institute of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China
- Department of Health Management Center & Institute of Health Management, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Zihan Li
- Tianjin Institute of Immunology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, School of Basic Medical Sciences, Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, Tianjin Medical University, Tianjin, China
- Department of Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin, China
- Department of Anesthesiology, Tianjin Institute of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Lina Ni
- Tianjin Institute of Immunology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, School of Basic Medical Sciences, Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, Tianjin Medical University, Tianjin, China
- Department of Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin, China
- Department of Anesthesiology, Tianjin Institute of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Sujun Yu
- Tianjin Institute of Immunology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, School of Basic Medical Sciences, Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, Tianjin Medical University, Tianjin, China
- Department of Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin, China
- Department of Anesthesiology, Tianjin Institute of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiao Shan
- Tianjin Institute of Immunology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, School of Basic Medical Sciences, Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, Tianjin Medical University, Tianjin, China
- Department of Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin, China
- Department of Anesthesiology, Tianjin Institute of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Penghui Hu
- Tianjin Institute of Immunology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, School of Basic Medical Sciences, Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, Tianjin Medical University, Tianjin, China
- Department of Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin, China
- Department of Anesthesiology, Tianjin Institute of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Zemin Ji
- Tianjin Institute of Immunology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, School of Basic Medical Sciences, Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, Tianjin Medical University, Tianjin, China
- Department of Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin, China
- Department of Anesthesiology, Tianjin Institute of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Weijia Jing
- Tianjin Institute of Immunology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, School of Basic Medical Sciences, Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, Tianjin Medical University, Tianjin, China
- Department of Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin, China
- Department of Anesthesiology, Tianjin Institute of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Yanzhao Zhou
- Tianjin Institute of Immunology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, School of Basic Medical Sciences, Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, Tianjin Medical University, Tianjin, China
- Department of Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin, China
- Department of Anesthesiology, Tianjin Institute of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Baochen Wang
- Tianjin Institute of Immunology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, School of Basic Medical Sciences, Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, Tianjin Medical University, Tianjin, China
- Department of Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin, China
- Department of Anesthesiology, Tianjin Institute of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Hongyuan Dong
- Tianjin Institute of Immunology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, School of Basic Medical Sciences, Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, Tianjin Medical University, Tianjin, China
- Department of Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin, China
- Department of Anesthesiology, Tianjin Institute of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Jinxue Zhou
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Keliang Xie
- Department of Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin, China
- Department of Anesthesiology, Tianjin Institute of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Qiujing Yu
- Tianjin Institute of Immunology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, School of Basic Medical Sciences, Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, Tianjin Medical University, Tianjin, China
- Department of Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin, China
- Department of Anesthesiology, Tianjin Institute of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China
- Department of Health Management Center & Institute of Health Management, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
105
|
Liu C, Ma N, Sun C, Shen X, Li J, Wang C. The effect of magnesium ions synergistic with mineralized collagen on osteogenesis/angiogenesis properties by modulating macrophage polarization in vitroand in vivo. Biomed Mater 2024; 19:035028. [PMID: 38518370 DOI: 10.1088/1748-605x/ad3702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 03/22/2024] [Indexed: 03/24/2024]
Abstract
In bone tissue engineering, the bone immunomodulatory properties of biomaterials are critical for bone regeneration, which is a synergistic process involving physiological activities like immune response, osteogenesis, and angiogenesis. The effect of the macrophage immune microenvironment on the osteogenesis and angiogenesis of various material extracts was examined in this experiment using Mg2+and Nano-hydroxyapatite/collagen (nHAC) in both a single application and a combined form. This studyin vitrorevealed that the two compounds combined significantly inhibited the NF-κB signaling pathway and reduced the release of inflammatory factors from macrophages when compared with the extraction phase alone. Additionally, by contributing to the polarization of macrophages towards the M2 type, the combined effects of the two materials can significantly improve osteogenesis/angiogenesis. The results ofin vivoexperiments confirmed that Mg2+/nHAC significantly promoted bone regeneration and angiogenesis. This study offers a promising method for enhancing bone graft material osseointegration.
Collapse
Affiliation(s)
- Chang Liu
- Department of Prosthodontics, Second Affiliated Hospital (Stomatological Hospital Affiliated) of Jinzhou Medical University, Jinzhou 121000, People's Republic of China
- Collaborative Innovation Center for Health Promotion of Children and Adolescents of Jinzhou Medical University, Jinzhou 121000, People's Republic of China
| | - Nan Ma
- Department of Prosthodontics, Second Affiliated Hospital (Stomatological Hospital Affiliated) of Jinzhou Medical University, Jinzhou 121000, People's Republic of China
- Collaborative Innovation Center for Health Promotion of Children and Adolescents of Jinzhou Medical University, Jinzhou 121000, People's Republic of China
| | - Changan Sun
- Department of Prosthodontics, Second Affiliated Hospital (Stomatological Hospital Affiliated) of Jinzhou Medical University, Jinzhou 121000, People's Republic of China
| | - Xuecheng Shen
- Department of Prosthodontics, Second Affiliated Hospital (Stomatological Hospital Affiliated) of Jinzhou Medical University, Jinzhou 121000, People's Republic of China
| | - Jinwei Li
- School of Basic Medicine, Jinzhou Medical University, Jinzhou 121000, People's Republic of China
| | - Chengyue Wang
- Department of Prosthodontics, Second Affiliated Hospital (Stomatological Hospital Affiliated) of Jinzhou Medical University, Jinzhou 121000, People's Republic of China
- Collaborative Innovation Center for Health Promotion of Children and Adolescents of Jinzhou Medical University, Jinzhou 121000, People's Republic of China
| |
Collapse
|
106
|
Wang B, Yang Y, Ye J, Han X, Yang L, Huang Y, Chao L. GRIM-19 deficiency promotes macrophage polarization to the M1 phenotype partly through glycolysis in unexplained recurrent spontaneous abortion†. Biol Reprod 2024; 110:739-749. [PMID: 38206868 DOI: 10.1093/biolre/ioae005] [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: 08/17/2023] [Revised: 12/07/2023] [Accepted: 01/05/2024] [Indexed: 01/13/2024] Open
Abstract
The occurrence of unexplained recurrent spontaneous abortion (URSA) is closely related to immune system disorders, however, the underlying mechanisms remain unclear. The purpose of this study was to investigate the expression of GRIM-19 in URSA and the possible pathogenesis of URSA according to macrophage polarization. Here, we showed that GRIM-19 was downregulated in the uterine decidual macrophages of patients with URSA and that GRIM-19 downregulation was accompanied by increased M1 macrophage polarization. Furthermore, the expression levels of glycolytic enzymes were substantially enhanced in the uterine decidual macrophages of URSA patients, and glycolysis in THP-1-derived macrophages was further enhanced by the downregulation of GRIM-19. Additionally, the increase of M1 macrophages resulting from the loss of GRIM-19 was significantly reversed in cells treated with 2-deoxy-D-glucose (2-DG, an inhibitor of glycolysis). To provide more direct evidence, GRIM-19 deficiency was shown to promote macrophage polarization to the M1 phenotype in GRIM-19+/- mouse uteri. Overall, our study provides evidence that GRIM-19 deficiency may play a role in regulating macrophage polarization in URSA, and that glycolysis may participate in this process.
Collapse
Affiliation(s)
- Bingyu Wang
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| | - Yang Yang
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| | - Jinwen Ye
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| | - Xiaojuan Han
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| | - Lin Yang
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| | - Yufei Huang
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| | - Lan Chao
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| |
Collapse
|
107
|
Luo M, Zhao F, Cheng H, Su M, Wang Y. Macrophage polarization: an important role in inflammatory diseases. Front Immunol 2024; 15:1352946. [PMID: 38660308 PMCID: PMC11039887 DOI: 10.3389/fimmu.2024.1352946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Accepted: 03/26/2024] [Indexed: 04/26/2024] Open
Abstract
Macrophages are crucial cells in the human body's innate immunity and are engaged in a variety of non-inflammatory reactions. Macrophages can develop into two kinds when stimulated by distinct internal environments: pro-inflammatory M1-like macrophages and anti-inflammatory M2-type macrophages. During inflammation, the two kinds of macrophages are activated alternatively, and maintaining a reasonably steady ratio is critical for maintaining homeostasis in vivo. M1 macrophages can induce inflammation, but M2 macrophages suppress it. The imbalance between the two kinds of macrophages will have a significant impact on the illness process. As a result, there are an increasing number of research being conducted on relieving or curing illnesses by altering the amount of macrophages. This review summarizes the role of macrophage polarization in various inflammatory diseases, including autoimmune diseases (RA, EAE, MS, AIH, IBD, CD), allergic diseases (allergic rhinitis, allergic dermatitis, allergic asthma), atherosclerosis, obesity and type 2 diabetes, metabolic homeostasis, and the compounds or drugs that have been discovered or applied to the treatment of these diseases by targeting macrophage polarization.
Collapse
Affiliation(s)
| | | | | | | | - Yuanmin Wang
- The Third Affiliated Hospital of Zunyi Medical University, The First People’s Hospital of Zunyi, Zunyi, Guizhou, China
| |
Collapse
|
108
|
Gao F, Deng S, Liu Y, Wu P, Huang L, Zhu F, Wei C, Yuan Y, Gui Y, Tian Y, Fan H, Wu H. Compound sophora decoction alleviates ulcerative colitis by regulating macrophage polarization through cGAS inhibition: network pharmacology and experimental validation. Aging (Albany NY) 2024; 16:6921-6936. [PMID: 38613801 PMCID: PMC11087132 DOI: 10.18632/aging.205734] [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: 11/24/2023] [Accepted: 03/18/2024] [Indexed: 04/15/2024]
Abstract
INTRODUCTION Ulcerative colitis (UC) is a refractory disease with complex pathogenesis, and its pathogenesis is not clear. The present study aimed to investigate the potential target and related mechanism of Compound Sophora Decoction (CSD) in treating UC. METHODS A network pharmacology approach predicted the components and targets of CSD to treat UC, and cell and animal experiments confirmed the findings of the approach and a new target for CSD treatment of UC. RESULTS A total of 155 potential targets were identified for CSD treatment of UC, with some related to macrophage polarization, such as nitric oxide synthase (NOS2), also known as inducible nitric oxide synthase (iNOS). GO and KEGG enrichment analysis indicated that oxidative stress response and multiple inflammatory signaling pathways such as TNF-α may play a significant role. In vitro experiments revealed that Interferon-stimulated DNA (ISD) interference can cause polarization imbalances in Raw 264.7 and bone marrow-derived macrophages (BMDMs). Flow cytometry demonstrated that polarization of macrophages in the intestine, spleen, and lymph nodes in vivo was also unbalanced after dextran sulfate sodium (DSS) modeling with pathological intestinal injury. Both in vitro and in vivo studies indicated that after inducing inflammation, the levels of macrophage polarization-related markers (iNOS and Arg1) and inflammation-related factors (CCL17, IL10, TNF-α, and CXCL10) changed, accompanied by increased expression of cGAS. However, CSD treatment based on inflammation can inhibit the expression of cGAS protein and mRNA, lower the level of inflammatory factors, promote the expression of anti-inflammatory factors, and regulate macrophage polarization. CONCLUSION We concluded that CSD alleviated DSS-induced UC by inhibiting cGAS, thus regulating macrophage polarization.
Collapse
Affiliation(s)
- Fei Gao
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Shuangjiao Deng
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yujin Liu
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Pengcheng Wu
- Wuhan Central Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Lifen Huang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Feng Zhu
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Chunzhu Wei
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yuyi Yuan
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yang Gui
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yushi Tian
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Heng Fan
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Hui Wu
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
109
|
Gao C, Nie Y. Etomidate inhibits tumor growth of glioblastoma by regulating M1 macrophage polarization. Metab Brain Dis 2024; 39:569-576. [PMID: 38300392 DOI: 10.1007/s11011-023-01335-y] [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: 05/24/2023] [Accepted: 12/04/2023] [Indexed: 02/02/2024]
Abstract
Glioblastoma (GBM) is a common primary central nervous system tumor. Although the multimodal integrated treatment for GBM has made great progress in recent years, the overall survival time of GBM is still short. Thus, novel treatments for GBM are worth further investigation and exploration. This study aimed to investigate the effects of etomidate on GBM tumor growth and the underlying mechanism. A xenograft tumor model was established and treated with etomidate to assess tumor growth. Immunohistochemistry (IHC) assay evaluated the positive rate of Ki67 cells in tumor tissues. Cell counting kit (CCK)-8 and EdU assays accessed the cell viability and proliferation. Immunofluorescence (IF) staining detected the distribution of macrophage markers in tumor tissues. The percentages of M1- and M2-like macrophages in tumor-associated macrophages (TAMs) and co-culture system (macrophages and GBM cells) were detected using flow cytometry. Macrophage polarization-related genes were measured using reverse transcription-quantitative polymerase chain reaction (RT-qPCR). Etomidate treatment inhibited the tumor growth, and increased the CD86+ cells but decreased the CD206+ cells in TAMs. The gene expression of M1 markers was increased in TAMs of etomidate-treated mice, whereas that of M2 markers was decreased. Moreover, etomidate treatment increased the number of CD86+ M1-like macrophages co-cultured with tumor cells but decreased that of CD206+ M2-like macrophages, with the upregulation of M1 markers and downregulation of M2 markers. Etomidate inhibited GBM tumor growth by promoting M1 macrophage polarization, suggesting a new insight into the clinical treatment of GBM.
Collapse
Affiliation(s)
- Caiyan Gao
- Department of Anesthesiology, The First Affiliated Hospital of Harbin Medical University, No. 23, Youzheng Street, Nangang District, Harbin, 150001, P.R. China
| | - Yan Nie
- Department of Anesthesiology, The First Affiliated Hospital of Harbin Medical University, No. 23, Youzheng Street, Nangang District, Harbin, 150001, P.R. China.
| |
Collapse
|
110
|
Bao S, Yu D, Tang Z, Wu H, Zhang H, Wang N, Liu Y, Huang H, Liu C, Li X, Guo Z. Conformationally regulated "nanozyme-like" cerium oxide with multiple free radical scavenging activities for osteoimmunology modulation and vascularized osseointegration. Bioact Mater 2024; 34:64-79. [PMID: 38186961 PMCID: PMC10770363 DOI: 10.1016/j.bioactmat.2023.12.006] [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: 10/07/2023] [Revised: 11/20/2023] [Accepted: 12/05/2023] [Indexed: 01/09/2024] Open
Abstract
Given post-operative aseptic loosening in orthopedic disease treatment, osteointegration occurs at the bone-implant interface as a holistic process, including immunoregulation (e.g., macrophage polarization), angiogenesis and osteogenesis in sequence. In order to achieve early rapid and satisfactory osseointegration, different nano-shaped (nanocone, nanopolyhedron and nanoflower abbr. NC, NP & NF) cerium oxide (CeO2-x) coatings, endowed with "nanozyme-like" activities for multiple free radical elimination and osteoimmunology regulation, were hydrothermally synthesized on titanium alloy (TC4). In vitro cell experiments showed that nano-CeO2-x coated TC4 not only induced polarization of RAW264.7 cells toward M2 phenotype, but also promoted angiogenesis and vascularization of endothelial cells along with differentiation and mineralization of osteogenic precursor cells. Improvements in M2-polarized macrophage, angiogenesis, and bone regeneration were further confirmed in a rat femoral condyle model. Among the above three nano-morphologies, NF exhibited the best osseoinetegration. RNA sequencing and mechanism exploration suggested that the inhibition of PI3K-AKT signaling pathway was essential for immunomodulatory capacity of NF. In conclusion, it provided promising insights into the immunomodulatory exploitation of orthopedic implants.
Collapse
Affiliation(s)
- Shusen Bao
- Department of Orthopedics, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China
- Department of Orthopedics, No. 903 Hospital of PLA Joint Logistic Support Force, Hangzhou, 310000, China
| | - Dongmei Yu
- Department of Orthopedics, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China
- University College London, UCL Institute of Orthopaedics and Musculo-Skeletal Science, M14 the Royal National Orthopaedic Hospital, Stanmore, HA7 4LP, United Kingdom
| | - Zhen Tang
- Department of Orthopedics, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China
| | - Hao Wu
- Department of Orthopedics, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China
| | - Hao Zhang
- Department of Burn Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710038, China
| | - Ning Wang
- Department of Orthopedics, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China
| | - Yichao Liu
- Department of Orthopedics, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China
| | - Hai Huang
- Department of Orthopedics, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China
| | - Chaozong Liu
- University College London, UCL Institute of Orthopaedics and Musculo-Skeletal Science, M14 the Royal National Orthopaedic Hospital, Stanmore, HA7 4LP, United Kingdom
| | - Xiaokang Li
- Department of Orthopedics, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China
| | - Zheng Guo
- Department of Orthopedics, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China
| |
Collapse
|
111
|
Povo-Retana A, Sánchez-García S, Alvarez-Lucena C, Landauro-Vera R, Prieto P, Delgado C, Martín-Sanz P, Boscá L. Crosstalk between P2Y receptors and cyclooxygenase activity in inflammation and tissue repair. Purinergic Signal 2024; 20:145-155. [PMID: 37052777 PMCID: PMC10997571 DOI: 10.1007/s11302-023-09938-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: 12/20/2022] [Accepted: 03/29/2023] [Indexed: 04/14/2023] Open
Abstract
The role of extracellular nucleotides as modulators of inflammation and cell stress is well established. One of the main actions of these molecules is mediated by the activation of purinergic receptors (P2) of the plasma membrane. P2 receptors can be classified according to two different structural families: P2X ionotropic ion channel receptors, and P2Y metabotropic G protein-coupled receptors. During inflammation, damaged cells release nucleotides and purinergic signaling occurs along the temporal pattern of the synthesis of pro-inflammatory and pro-resolving mediators by myeloid and lymphoid cells. In macrophages under pro-inflammatory conditions, the expression and activity of cyclooxygenase 2 significantly increases and enhances the circulating levels of prostaglandin E2 (PGE2), which exerts its effects both through specific plasma membrane receptors (EP1-EP4) and by activation of intracellular targets. Here we review the mechanisms involved in the crosstalk between PGE2 and P2Y receptors on macrophages, which is dependent on several isoforms of protein kinase C and protein kinase D1. Due to this crosstalk, a P2Y-dependent increase in calcium is blunted by PGE2 whereas, under these conditions, macrophages exhibit reduced migratory capacity along with enhanced phagocytosis, which contributes to the modulation of the inflammatory response and tissue repair.
Collapse
Affiliation(s)
- Adrián Povo-Retana
- Instituto de Investigaciones Biomédicas Alberto Sols (Centro Mixto CSIC-UAM), Arturo Duperier 4, 28029, Madrid, Spain.
| | - Sergio Sánchez-García
- Instituto de Investigaciones Biomédicas Alberto Sols (Centro Mixto CSIC-UAM), Arturo Duperier 4, 28029, Madrid, Spain
| | - Carlota Alvarez-Lucena
- Instituto de Investigaciones Biomédicas Alberto Sols (Centro Mixto CSIC-UAM), Arturo Duperier 4, 28029, Madrid, Spain
| | - Rodrigo Landauro-Vera
- Instituto de Investigaciones Biomédicas Alberto Sols (Centro Mixto CSIC-UAM), Arturo Duperier 4, 28029, Madrid, Spain
| | - Patricia Prieto
- Departamento de Farmacología, Farmacognosia y Botánica. Facultad de Farmacia, Universidad Complutense de Madrid, Plaza Ramón y Cajal, 28040, Madrid, Spain
| | - Carmen Delgado
- Instituto de Investigaciones Biomédicas Alberto Sols (Centro Mixto CSIC-UAM), Arturo Duperier 4, 28029, Madrid, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Melchor Fernández Almagro 6, 28029, Madrid, Spain
| | - Paloma Martín-Sanz
- Instituto de Investigaciones Biomédicas Alberto Sols (Centro Mixto CSIC-UAM), Arturo Duperier 4, 28029, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Melchor Fernández Almagro 6, 28029, Madrid, Spain
| | - Lisardo Boscá
- Instituto de Investigaciones Biomédicas Alberto Sols (Centro Mixto CSIC-UAM), Arturo Duperier 4, 28029, Madrid, Spain.
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Melchor Fernández Almagro 6, 28029, Madrid, Spain.
| |
Collapse
|
112
|
Ratanasereeprasert N, Hsu LF, Wang SK, Chen YJ, Chang JH, Yao CCJ. Molecular signaling and mechanisms of low-level laser-induced gene expression in cells involved in orthodontic tooth movement. J Formos Med Assoc 2024; 123:442-451. [PMID: 37805307 DOI: 10.1016/j.jfma.2023.09.011] [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: 04/25/2023] [Revised: 08/03/2023] [Accepted: 09/19/2023] [Indexed: 10/09/2023] Open
Abstract
BACKGROUND The study aimed to observe molecular signaling, including reactive oxygen species (ROS) and mitochondrial membrane potential (ΔΨm), to evaluate the alteration of gene expression by low-level laser therapy (LLLT) and the correlation between its mechanisms and the NF-kB pathway in cells involved in orthodontic tooth movement. METHODS Osteoblast-like cells (MG63), immortalized periodontal ligament cells (iPDL), and M1 macrophage-like cells were irradiated by 980-nm LLLT with energy densities of 1 and 10 J/cm2 ΔΨm and intracellular ROS were monitored using fluorescent probes. The changes of mRNA expression were assessed using reverse transcription polymerase chain reaction (RT-PCR). NF-kB inhibitor, ROS scavenger, and ΔΨm suppressor were used to analyze signals associated with the regulation of gene expression. Finally, Western blot analysis was performed to confirm NF-kB signaling after LLLT. RESULTS We found the increases of ΔΨm and ROS in all three cell types after LLLT, but no significant difference was observed between 1 and 10 J/cm2 LLLT. Regarding gene expression, some target genes were upregulated in MG63 6 h, 12 h, and 1 day after LLLT and in iPDL cells 12 h and 1 day after LLLT. However, no changes occurred in M1 cells. The inhibitor that significantly reduced most changes in gene expression was NF-kB inhibitor. Western blot analysis showed the increase in p-IkBα level after LLLT in iPDL and MG63, but not in M1. CONCLUSION The 980-nm LLLT increased ΔΨm and ROS production in all three cell types. However, changes in gene regulation were found only in MG63 and iPDL cells, which related to the NF-kB pathway.
Collapse
Affiliation(s)
| | - Li-Fang Hsu
- Department of Dentistry, National Taiwan University Hospital, Hsin-Chu Branch, Hsin-Chu, Taiwan
| | - Shih-Kai Wang
- Department of Dentistry, School of Dentistry, National Taiwan University, Department of Pediatric Dentistry, National Taiwan University Children's Hospital, Taipei, Taiwan
| | - Yi-Jane Chen
- Graduate Institute of Clinical Dentistry, School of Dentistry, National Taiwan University, Taipei, Taiwan; Division of Orthodontics and Dentofacial Orthopedics, Dental Department, National Taiwan University Hospital, Taipei, Taiwan
| | - Jui-Heng Chang
- Graduate Institute of Clinical Dentistry, School of Dentistry, National Taiwan University, Taipei, Taiwan
| | - Chung-Chen Jane Yao
- Graduate Institute of Clinical Dentistry, School of Dentistry, National Taiwan University, Taipei, Taiwan; Division of Orthodontics and Dentofacial Orthopedics, Dental Department, National Taiwan University Hospital, Taipei, Taiwan.
| |
Collapse
|
113
|
Wang J, Wu Z, Huang Y, Jin L, Xu J, Yao Z, Ouyang X, Zhou Z, Mao S, Cao J, Lai B, Shen W. IRF4 induces M1 macrophage polarization and aggravates ulcerative colitis progression by the Bcl6-dependent STAT3 pathway. ENVIRONMENTAL TOXICOLOGY 2024; 39:2390-2404. [PMID: 38164749 DOI: 10.1002/tox.24106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 12/05/2023] [Accepted: 12/10/2023] [Indexed: 01/03/2024]
Abstract
Ulcerative colitis (UC) is an idiopathic chronic intestinal inflammation. An increasing body of evidence shows that macrophages play an important role in the pathogenesis of UC. Interferon regulatory factor 4 (IRF4) is crucial for the development of autoimmune diseases via regulating immune cells. This research was designed to explore the function of IRF4 in UC and its association with macrophage polarization. The in vitro model of UC was established by stimulating colonic epithelial cells with tumor necrosis factor α (TNF-α). A mouse model of UC was constructed by injecting C57BL/6 mice with dextran sulfate sodium salt. Flow cytometry was used to assess percentage of CD11b+ CD86+ and CD11b+ CD206+ cells in bone marrow macrophages. Occult blood tests were used to detect hematochezia. Hematoxylin and eosin staining assay was used to assess colon pathological changes. Enzyme-linked immunosorbent assay (ELISA) was used to detect concentrations of inflammatory cytokines. The interaction of IRF4 and B-cell lymphoma 6 (Bcl6) was confirmed using GST pull-down and coimmunoprecipitation assays. Our findings revealed that IRF4 promoted cell apoptosis and stimulated M1 macrophage polarization in vitro. Furthermore, IRF4 aggravated symptoms of the mouse model of UC and aggravated M1 macrophage polarization in vivo. IRF4 negatively regulated Bcl6 expression. Downregulation of Bcl6 promoted apoptosis and M1 macrophage polarization in the presence of IRF4 in vitro and in vivo. Moreover, Bcl6 positively mediated the Janus kinase 2 (JAK2)/signal transducer and activator of transcription 3 (STAT3) signaling pathway. In conclusion, IRF4 aggravated UC progression through promoting M1 macrophage polarization via Bcl6/JAK2/STAT3 pathway. These findings suggested that IRF4 might be a good target to competitively inhibit or to treat with UC.
Collapse
Affiliation(s)
- Jiwei Wang
- Department of Ultrasound, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Zhao Wu
- Nanchang University, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Yulin Huang
- Nanchang University, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Lin Jin
- Nanchang University, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Jinyi Xu
- Nanchang University, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Zhiyi Yao
- Nanchang University, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Xi Ouyang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Zhiyong Zhou
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Shengxun Mao
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Jiaqing Cao
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Bin Lai
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Wei Shen
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
114
|
Cao Y, Sun H, Li X, Pommer W, Xiong Y, Chen X, Chu C, Yu F, Hocher B, Wang Z. GSK343 modulates macrophage M2 polarization through the EZH2/MST1/YAP1 signaling axis to mitigate neurological damage induced by hypercalcemia in CKD mice. Cell Signal 2024; 116:111063. [PMID: 38242267 DOI: 10.1016/j.cellsig.2024.111063] [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: 01/11/2024] [Accepted: 01/16/2024] [Indexed: 01/21/2024]
Abstract
Chronic kidney disease (CKD) often culminates in hypercalcemia, instigating severe neurological injuries that are not yet fully understood. This study unveils a mechanism, where GSK343 ameliorates CKD-induced neural damage in mice by modulating macrophage polarization through the EZH2/MST1/YAP1 signaling axis. Specifically, GSK343 downregulated the expression of histone methyltransferase EZH2 and upregulated MST1, which suppressed YAP1, promoting M2 macrophage polarization and thereby, alleviating neural injury in hypercalcemia arising from renal failure. This molecular pathway introduced herein not only sheds light on the cellular machinations behind CKD-induced neurological harm but also paves the way for potential therapeutic interventions targeting the identified axis, especially considering the M2 macrophage polarization as a potential strategy to mitigate hypercalcemia-induced neural injuries.
Collapse
Affiliation(s)
- Yaochen Cao
- Department of Medicine, Hainan Medical University, The First Affiliated Hospital of Hainan Medical University, No. 31 Longhua Road, Haikou 570102, Hannan, China; Department of Nephrology, Charité - Universitätsmedizin Berlin, Campus Mitte, Berlin 10117, Germany; Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany; Key Laboratory of Emergency and Trauma, Ministry of Education, Key Laboratory of Hainan Trauma and Disaster Rescue, The First Affiliated Hospital of Hainan Medical University, College of Pharmacy, Institute of Functional Materials and Molecular Imaging, College of Emergency and Trauma, Hainan Medical University, Haikou 571199, China
| | - Hongming Sun
- Department of Medicine, Hainan Medical University, The First Affiliated Hospital of Hainan Medical University, No. 31 Longhua Road, Haikou 570102, Hannan, China; Department of Neurology and Neuroscience, Okayama University School of Medicine, Okayama 700-8558, Japan
| | - Xitong Li
- Department of Nephrology, Charité - Universitätsmedizin Berlin, Campus Mitte, Berlin 10117, Germany; Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Wolfgang Pommer
- Kuratorium für Dialyse und Nierentransplantation (KfH) - Bildungszentrum, Martin-Behaim-Str. 20, Neu-Isenburg 63263, Germany
| | - Yingquan Xiong
- Department of Nephrology, Charité - Universitätsmedizin Berlin, Campus Mitte, Berlin 10117, Germany; Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Xin Chen
- Department of Nephrology, Charité - Universitätsmedizin Berlin, Campus Mitte, Berlin 10117, Germany; Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Chang Chu
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Fabiao Yu
- Department of Medicine, Hainan Medical University, The First Affiliated Hospital of Hainan Medical University, No. 31 Longhua Road, Haikou 570102, Hannan, China; Key Laboratory of Emergency and Trauma, Ministry of Education, Key Laboratory of Hainan Trauma and Disaster Rescue, The First Affiliated Hospital of Hainan Medical University, College of Pharmacy, Institute of Functional Materials and Molecular Imaging, College of Emergency and Trauma, Hainan Medical University, Haikou 571199, China.
| | - Berthold Hocher
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany; Key Laboratory of Emergency and Trauma, Ministry of Education, Key Laboratory of Hainan Trauma and Disaster Rescue, The First Affiliated Hospital of Hainan Medical University, College of Pharmacy, Institute of Functional Materials and Molecular Imaging, College of Emergency and Trauma, Hainan Medical University, Haikou 571199, China; Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, China; Institute of Medical Diagnostics, IMD Berlin, Berlin, Germany.
| | - Ziqiang Wang
- Department of Medicine, Hainan Medical University, The First Affiliated Hospital of Hainan Medical University, No. 31 Longhua Road, Haikou 570102, Hannan, China; Key Laboratory of Emergency and Trauma, Ministry of Education, Key Laboratory of Hainan Trauma and Disaster Rescue, The First Affiliated Hospital of Hainan Medical University, College of Pharmacy, Institute of Functional Materials and Molecular Imaging, College of Emergency and Trauma, Hainan Medical University, Haikou 571199, China.
| |
Collapse
|
115
|
Cai L, Xia M, Zhang F. Redox Regulation of Immunometabolism in Microglia Underpinning Diabetic Retinopathy. Antioxidants (Basel) 2024; 13:423. [PMID: 38671871 PMCID: PMC11047590 DOI: 10.3390/antiox13040423] [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: 01/31/2024] [Revised: 03/24/2024] [Accepted: 03/28/2024] [Indexed: 04/28/2024] Open
Abstract
Diabetic retinopathy (DR) is the leading cause of visual impairment and blindness among the working-age population. Microglia, resident immune cells in the retina, are recognized as crucial drivers in the DR process. Microglia activation is a tightly regulated immunometabolic process. In the early stages of DR, the M1 phenotype commonly shifts from oxidative phosphorylation to aerobic glycolysis for energy production. Emerging evidence suggests that microglia in DR not only engage specific metabolic pathways but also rearrange their oxidation-reduction (redox) system. This redox adaptation supports metabolic reprogramming and offers potential therapeutic strategies using antioxidants. Here, we provide an overview of recent insights into the involvement of reactive oxygen species and the distinct roles played by key cellular antioxidant pathways, including the NADPH oxidase 2 system, which promotes glycolysis via enhanced glucose transporter 4 translocation to the cell membrane through the AKT/mTOR pathway, as well as the involvement of the thioredoxin and nuclear factor E2-related factor 2 antioxidant systems, which maintain microglia in an anti-inflammatory state. Therefore, we highlight the potential for targeting the modulation of microglial redox metabolism to offer new concepts for DR treatment.
Collapse
Affiliation(s)
- Luwei Cai
- National Clinical Research Center for Eye Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China; (L.C.); (M.X.)
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai 200080, China
- Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai 200080, China
- Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai 200080, China
| | - Mengxue Xia
- National Clinical Research Center for Eye Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China; (L.C.); (M.X.)
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai 200080, China
- Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai 200080, China
- Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai 200080, China
| | - Fang Zhang
- National Clinical Research Center for Eye Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China; (L.C.); (M.X.)
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai 200080, China
- Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai 200080, China
- Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai 200080, China
| |
Collapse
|
116
|
Soldano S, Smith V, Montagna P, Gotelli E, Campitiello R, Pizzorni C, Paolino S, Sulli A, Cere A, Cutolo M. Nintedanib downregulates the profibrotic M2 phenotype in cultured monocyte-derived macrophages obtained from systemic sclerosis patients affected by interstitial lung disease. Arthritis Res Ther 2024; 26:74. [PMID: 38509595 PMCID: PMC10953168 DOI: 10.1186/s13075-024-03308-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 03/10/2024] [Indexed: 03/22/2024] Open
Abstract
BACKGROUND Systemic sclerosis (SSc) is an autoimmune connective tissue disease characterized by vasculopathy and progressive fibrosis of skin and several internal organs, including lungs. Macrophages are the main cells involved in the immune-inflammatory damage of skin and lungs, and alternatively activated (M2) macrophages seem to have a profibrotic role through the release of profibrotic cytokines (IL10) and growth factors (TGFβ1). Nintedanib is a tyrosine kinase inhibitor targeting several fibrotic mediators and it is approved for the treatment of SSc-related interstitial lung disease (ILD). The study aimed to evaluate the effect of nintedanib in downregulating the profibrotic M2 phenotype in cultured monocyte-derived macrophages (MDMs) obtained from SSc-ILD patients. METHODS Fourteen SSc patients, fulfilling the 2013 ACR/EULAR criteria for SSc, 10 SSc patients affected by ILD (SSc-ILD pts), 4 SSc patients non affected by ILD (SSc pts no-ILD), and 5 voluntary healthy subjects (HSs), were recruited at the Division of Clinical Rheumatology-University of Genova, after obtaining Ethical Committee approval and patients' informed consent. Monocytes were isolated from peripheral blood, differentiated into MDMs, and then maintained in growth medium without any treatment (untreated cells), or treated with nintedanib (0.1 and 1µM) for 3, 16, and 24 h. Gene expression of macrophage scavenger receptors (CD204, CD163), mannose receptor-1 (CD206), Mer tyrosine kinase (MerTK), identifying M2 macrophages, together with TGFβ1 and IL10, were evaluated by quantitative real-time polymerase chain reaction. Protein synthesis was investigated by Western blotting and the level of active TGFβ1 was evaluated by ELISA. Statistical analysis was carried out using non-parametric Wilcoxon test. RESULTS Cultured untreated SSc-ILD MDMs showed a significant increased protein synthesis of CD206 (p < 0.05), CD204, and MerTK (p < 0.01), together with a significant upregulation of the gene expression of MerTK and TGFβ1 (p < 0.05; p < 0.01) compared to HS-MDMs. Moreover, the protein synthesis of CD206 and MerTK and the gene expression of TGFβ1 were significantly higher in cultured untreated MDMs from SSc-ILD pts compared to MDMs without ILD (p < 0.05; p < 0.01). In cultured SSc-ILD MDMs, nintedanib 0.1 and 1µM significantly downregulated the gene expression and protein synthesis of CD204, CD206, CD163 (p < 0.05), and MerTK (p < 0.01) compared to untreated cells after 24 h of treatment. Limited to MerTK and IL10, both nintedanib concentrations significantly downregulated their gene expression already after 16 h of treatment (p < 0.05). In cultured SSc-ILD MDMs, nintedanib 0.1 and 1µM significantly reduced the release of active TGFβ1 after 24 h of treatment (p < 0.05 vs. untreated cells). CONCLUSIONS In cultured MDMs from SSc-ILD pts, nintedanib seems to downregulate the profibrotic M2 phenotype through the significant reduction of gene expression and protein synthesis of M2 cell surface markers, together with the significant reduction of TGFβ1 release, and notably MerTK, a tyrosine kinase receptor involved in lung fibrosis.
Collapse
Affiliation(s)
- Stefano Soldano
- Laboratory of Experimental Rheumatology, Division of Clinical Rheumatology, Department of Internal Medicine, University of Genova, Genoa, Italy
| | - Vanessa Smith
- Department of Internal Medicine, Ghent University, Ghent, Belgium
- Department of Rheumatology, Ghent University Hospital, Ghent, Belgium
- Unit for Molecular Immunology and Inflammation, VIB Inflammation Research Centre, Ghent, Belgium
| | - Paola Montagna
- Laboratory of Experimental Rheumatology, Division of Clinical Rheumatology, Department of Internal Medicine, University of Genova, Genoa, Italy
| | - Emanuele Gotelli
- Laboratory of Experimental Rheumatology, Division of Clinical Rheumatology, Department of Internal Medicine, University of Genova, Genoa, Italy
| | - Rosanna Campitiello
- Laboratory of Experimental Rheumatology, Division of Clinical Rheumatology, Department of Internal Medicine, University of Genova, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Carmen Pizzorni
- Laboratory of Experimental Rheumatology, Division of Clinical Rheumatology, Department of Internal Medicine, University of Genova, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Sabrina Paolino
- Laboratory of Experimental Rheumatology, Division of Clinical Rheumatology, Department of Internal Medicine, University of Genova, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Alberto Sulli
- Laboratory of Experimental Rheumatology, Division of Clinical Rheumatology, Department of Internal Medicine, University of Genova, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Andrea Cere
- Laboratory of Experimental Rheumatology, Division of Clinical Rheumatology, Department of Internal Medicine, University of Genova, Genoa, Italy
| | - Maurizio Cutolo
- Laboratory of Experimental Rheumatology, Division of Clinical Rheumatology, Department of Internal Medicine, University of Genova, Genoa, Italy.
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy.
| |
Collapse
|
117
|
Sadeghi M, Dehnavi S, Sharifat M, Amiri AM, Khodadadi A. Innate immune cells: Key players of orchestra in modulating tumor microenvironment (TME). Heliyon 2024; 10:e27480. [PMID: 38463798 PMCID: PMC10923864 DOI: 10.1016/j.heliyon.2024.e27480] [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: 11/06/2023] [Revised: 02/29/2024] [Accepted: 02/29/2024] [Indexed: 03/12/2024] Open
Abstract
The tumor microenvironment (TME) with vital role in cancer progression is composed of various cells such as endothelial cells, immune cells, and mesenchymal stem cells. In particular, innate immune cells such as macrophages, dendritic cells, myeloid-derived suppressor cells, neutrophils, innate lymphoid cells, γδT lymphocytes, and natural killer cells can either promote or suppress tumor progression when present in the TME. An increase in research on the cross-talk between the TME and innate immune cells will lead to new approaches for anti-tumoral therapeutic interventions. This review primarily focuses on the biology of innate immune cells and their main functions in the TME. In addition, it summarizes several innate immune-based immunotherapies that are currently tested in clinical trials.
Collapse
Affiliation(s)
- Mahvash Sadeghi
- Department of Immunology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Student Research Committee, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Sajad Dehnavi
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Moosa Sharifat
- Department of Immunology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Amir Mohammad Amiri
- Department of Immunology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Student Research Committee, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Ali Khodadadi
- Department of Immunology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Cancer, Petroleum and Environmental Pollutants Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
118
|
Lou K, Luo H, Jiang X, Feng S. Applications of emerging extracellular vesicles technologies in the treatment of inflammatory diseases. Front Immunol 2024; 15:1364401. [PMID: 38545101 PMCID: PMC10965547 DOI: 10.3389/fimmu.2024.1364401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 03/04/2024] [Indexed: 04/17/2024] Open
Abstract
The emerging extracellular vesicles technologies is an advanced therapeutic approach showing promising potential for addressing inflammatory diseases. These techniques have been proven to have positive effects on immune modulation and anti-inflammatory responses. With these advancements, a comprehensive review and update on the role of extracellular vesicles in inflammatory diseases have become timely. This review aims to summarize the research progress of extracellular vesicle technologies such as plant-derived extracellular vesicles, milk-derived extracellular vesicles, mesenchymal stem cell-derived extracellular vesicles, macrophage-derived extracellular vesicles, etc., in the treatment of inflammatory diseases. It elucidates their potential significance in regulating inflammation, promoting tissue repair, and treating diseases. The goal is to provide insights for future research in this field, fostering the application and development of extracellular vesicle technology in the treatment of inflammatory diseases.
Collapse
Affiliation(s)
- Kecheng Lou
- Department of Urology, Lanxi People’s Hospital, Jinhua, Zhejiang, China
| | - Hui Luo
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Xinghua Jiang
- Department of Urology, Jingdezhen Second People’s Hospital, Jingdezhen, Jiangxi, China
| | - Shangzhi Feng
- Department of Urology, Jiujiang University Clinic College/Hospital, Jiujiang, Jiangxi, China
| |
Collapse
|
119
|
Zhou BW, Liu HM, Xu F, Jia XH. The role of macrophage polarization and cellular crosstalk in the pulmonary fibrotic microenvironment: a review. Cell Commun Signal 2024; 22:172. [PMID: 38461312 PMCID: PMC10924385 DOI: 10.1186/s12964-024-01557-2] [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: 12/28/2023] [Accepted: 03/03/2024] [Indexed: 03/11/2024] Open
Abstract
Pulmonary fibrosis (PF) is a progressive interstitial inflammatory disease with a high mortality rate. Patients with PF commonly experience a chronic dry cough and progressive dyspnoea for years without effective mitigation. The pathogenesis of PF is believed to be associated with dysfunctional macrophage polarization, fibroblast proliferation, and the loss of epithelial cells. Thus, it is of great importance and necessity to explore the interactions among macrophages, fibroblasts, and alveolar epithelial cells in lung fibrosis, as well as in the pro-fibrotic microenvironment. In this review, we discuss the latest studies that have investigated macrophage polarization and activation of non-immune cells in the context of PF pathogenesis and progression. Next, we discuss how profibrotic cellular crosstalk is promoted in the PF microenvironment by multiple cytokines, chemokines, and signalling pathways. And finally, we discuss the potential mechanisms of fibrogenesis development and efficient therapeutic strategies for the disease. Herein, we provide a comprehensive summary of the vital role of macrophage polarization in PF and its profibrotic crosstalk with fibroblasts and alveolar epithelial cells and suggest potential treatment strategies to target their cellular communication in the microenvironment.
Collapse
Affiliation(s)
- Bo-Wen Zhou
- The First School of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Hua-Man Liu
- Department of General Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Fei Xu
- Department of Pneumology and Critical Care Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Xin-Hua Jia
- Department of Pneumology and Critical Care Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China.
| |
Collapse
|
120
|
Dai Y, Yi X, Huang Y, Qian K, Huang L, Hu J, Liu Y. miR-345-3p Modulates M1/M2 Macrophage Polarization to Inhibit Inflammation in Bone Infection via Targeting MAP3K1 and NF-κB Pathway. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:844-854. [PMID: 38231123 DOI: 10.4049/jimmunol.2300561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 12/20/2023] [Indexed: 01/18/2024]
Abstract
Infection after fracture fixation (IAFF), a complex infectious disease, causes inflammatory destruction of bone tissue and poses a significant clinical challenge. miR-345-3p is a biomarker for tibial infected nonunion; however, the comprehensive mechanistic role of miR-345-3p in IAFF is elusive. In this study, we investigated the role of miR-345-3p in IAFF pathogenesis through in vivo and in vitro experiments. In vivo, in a rat model of IAFF, miR-345-3p expression was downregulated, accompanied by increased M1 macrophage infiltration and secretion of proinflammatory factors. In vitro, LPS induced differentiation of primary rat bone marrow-derived macrophages into M1 macrophages, which was attenuated by miR-345-3p mimics. miR-345-3p promoted M1 to M2 macrophage transition-it reduced the expression of cluster of differentiation (CD) 86, inducible NO synthase, IL-1β, and TNF-α but elevated those of CD163, arginase-1, IL-4, and IL-10. MAPK kinase kinase 1 (MAP3K1), a target mRNA of miR-345-3p, was overexpressed in the bone tissue of IAFF rats compared with that in those of the control rats. The M1 to M2 polarization inhibited MAP3K1 signaling pathways in vitro. Conversely, MAP3K1 overexpression promoted the transition from M2 to M1. miR-345-3p significantly inhibited NF-κB translocation from the cytosol to the nucleus in a MAP3K1-dependent manner. In conclusion, miR-345-3p promotes the polarization of M1 macrophages to the M2 phenotype by inhibiting the MAP3K1 and NF-κB pathways. These findings provide insight into the pathogenesis and immunotherapeutic strategies for IAFF and offer potential new targets for subsequent research.
Collapse
Affiliation(s)
- Yan Dai
- Department of Infectious Diseases, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiaolan Yi
- Department of Infectious Diseases, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yahui Huang
- Department of Infectious Diseases, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Kaoliang Qian
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Lili Huang
- Department of Infectious Diseases, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jun Hu
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yuan Liu
- Department of Infectious Diseases, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
121
|
Wang H, Wang X, Zhang X, Xu W. The promising role of tumor-associated macrophages in the treatment of cancer. Drug Resist Updat 2024; 73:101041. [PMID: 38198845 DOI: 10.1016/j.drup.2023.101041] [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: 10/07/2023] [Revised: 12/16/2023] [Accepted: 12/28/2023] [Indexed: 01/12/2024]
Abstract
Macrophages are important components of the immune system. Mature macrophages can be recruited to tumor microenvironment that affect tumor cell proliferation, invasion and metastasis, extracellular matrix remodeling, immune suppression, as well as chemotherapy resistance. Classically activated type I macrophages (M1) exhibited marked tumor killing and phagocytosis. Therefore, using macrophages for adoptive cell therapy has attracted attention and become one of the most effective strategies for cancer treatment. Through cytokines and/or chemokines, macrophage can inhibit myeloid cells recruitment, and activate anti-tumor and immune killing functions. Applying macrophages for anti-tumor delivery is one of the most promising approaches for cancer therapy. This review article introduces the role of macrophages in tumor development and drug resistance, and the possible clinical application of targeting macrophages for overcoming drug resistance and enhancing cancer therapeutics, as well as its challenges.
Collapse
Affiliation(s)
- Hongbin Wang
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Harbin Medical University, PR China; Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin Medical University, PR China; Department of Surgical Oncology, Harbin Medical University Cancer Hospital, PR China.
| | - Xueying Wang
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, PR China; Otolaryngology Major Disease Research Key Laboratory of Hunan Province, PR China
| | - Xin Zhang
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, PR China; Otolaryngology Major Disease Research Key Laboratory of Hunan Province, PR China
| | - Wanhai Xu
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Harbin Medical University, PR China; Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin Medical University, PR China; Department of Urology, Harbin Medical University Cancer Hospital, PR China.
| |
Collapse
|
122
|
Bahiraii S, Brenner M, Weckwerth W, Heiss EH. Sulforaphane impedes mitochondrial reprogramming and histone acetylation in polarizing M1 (LPS) macrophages. Free Radic Biol Med 2024; 213:443-456. [PMID: 38301976 DOI: 10.1016/j.freeradbiomed.2024.01.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 01/18/2024] [Indexed: 02/03/2024]
Abstract
M1 (LPS) macrophages are characterized by a high expression of pro-inflammatory mediators, and distinct metabolic features that comprise increased glycolysis, a broken TCA cycle, or impaired OXPHOS with augmented mitochondrial ROS production. This study investigated whether the phytochemical sulforaphane (Sfn) influences mitochondrial reprogramming during M1 polarization, as well as to what extent this can contribute to Sfn-mediated inhibition of M1 marker expression in murine macrophages. The use of extracellular flux-, metabolite-, and immunoblot analyses as well as fluorescent dyes indicative for mitochondrial morphology, membrane potential or superoxide production, demonstrated that M1 (LPS/Sfn) macrophages maintain an unbroken TCA cycle, higher OXPHOS rate, boosted fusion dynamics, lower membrane potential, and less superoxide production in their mitochondria when compared to control M1 (LPS) cells. Sustained OXPHOS and TCA activity but not the concomitantly observed high dependency on fatty acids as fuel appeared necessary for M1 (LPS/Sfn) macrophages to reduce expression of nos2, il1β, il6 and tnfα. M1 (LPS/Sfn) macrophages also displayed lower nucleo/cytosolic acetyl-CoA levels in association with lower global and site-specific histone acetylation at selected pro-inflammatory gene promoters than M1 (LPS), evident in colorimetric coupled enzyme assays, immunoblot and ChIP-qPCR analyses, respectively. Supplementation with acetate or citrate was able to rescue both histone acetylation and mRNA expression of the investigated M1 marker genes in Sfn-treated cells. Overall, Sfn preserves mitochondrial functionality and restricts indispensable nuclear acetyl-CoA for histone acetylation and M1 marker expression in LPS-stimulated macrophages.
Collapse
Affiliation(s)
- Sheyda Bahiraii
- Department of Pharmaceutical Sciences, University of Vienna, Vienna, Austria; ViennaDoctoral School of Pharmaceutical, Nutritional and Sport Sciences (VDS PhaNuSpo), University of Vienna, Vienna, Austria
| | - Martin Brenner
- Department of Pharmaceutical Sciences, University of Vienna, Vienna, Austria; ViennaDoctoral School of Pharmaceutical, Nutritional and Sport Sciences (VDS PhaNuSpo), University of Vienna, Vienna, Austria; Vienna Metabolomics Center (VIME), University of Vienna, Vienna, Austria
| | - Wolfram Weckwerth
- Vienna Metabolomics Center (VIME), University of Vienna, Vienna, Austria; Molecular Systems Biology (MOSYS), Department of Functional and Evolutionary Ecology (FEE), University of Vienna, Vienna, Austria
| | - Elke H Heiss
- Department of Pharmaceutical Sciences, University of Vienna, Vienna, Austria.
| |
Collapse
|
123
|
Carvell T, Burgoyne P, Milne L, Campbell JDM, Fraser AR, Bridle H. Human leucocytes processed by fast-rate inertial microfluidics retain conventional functional characteristics. J R Soc Interface 2024; 21:20230572. [PMID: 38442860 PMCID: PMC10914517 DOI: 10.1098/rsif.2023.0572] [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: 10/02/2023] [Accepted: 02/01/2024] [Indexed: 03/07/2024] Open
Abstract
The manufacturing of clinical cellular therapies is a complex process frequently requiring manipulation of cells, exchange of buffers and volume reduction. Current manufacturing processes rely on either low throughput open centrifugation-based devices, or expensive closed-process alternatives. Inertial focusing (IF) microfluidic devices offer the potential for high-throughput, inexpensive equipment which can be integrated into a closed system, but to date no IF devices have been approved for use in cell therapy manufacturing, and there is limited evidence for the effects that IF processing has on human cells. The IF device described in this study was designed to simultaneously separate leucocytes, perform buffer exchange and provide a volume reduction to the cell suspension, using high flow rates with high Reynolds numbers. The performance and effects of the IF device were characterized using peripheral blood mononuclear cells and isolated monocytes. Post-processing cell effects were investigated using multi-parameter flow cytometry to track cell viability, functional changes and fate. The IF device was highly efficient at separating CD14+ monocytes (approx. 97% to one outlet, approx. 60% buffer exchange, 15 ml min-1) and leucocyte processing was well tolerated with no significant differences in downstream viability, immunophenotype or metabolic activity when compared with leucocytes processed with conventional processing techniques. This detailed approach provides robust evidence that IF devices could offer significant benefits to clinical cell therapy manufacture.
Collapse
Affiliation(s)
- Tom Carvell
- Institute of Biological Chemistry, Biophysics and Bioengineering, School of Engineering and Physical Sciences, Heriot-Watt University, Heriot-Watt Research Park, Edinburgh EH14 4AS, UK
| | - Paul Burgoyne
- Tissues, Cells and Advanced Therapeutics, Jack Copland Centre, Scottish National Blood Transfusion Service, Research Avenue North, Heriot-Watt Research Park, Edinburgh EH14 4BE, UK
| | - Laura Milne
- Tissues, Cells and Advanced Therapeutics, Jack Copland Centre, Scottish National Blood Transfusion Service, Research Avenue North, Heriot-Watt Research Park, Edinburgh EH14 4BE, UK
| | - John D. M. Campbell
- Tissues, Cells and Advanced Therapeutics, Jack Copland Centre, Scottish National Blood Transfusion Service, Research Avenue North, Heriot-Watt Research Park, Edinburgh EH14 4BE, UK
| | - Alasdair R. Fraser
- Tissues, Cells and Advanced Therapeutics, Jack Copland Centre, Scottish National Blood Transfusion Service, Research Avenue North, Heriot-Watt Research Park, Edinburgh EH14 4BE, UK
| | - Helen Bridle
- Institute of Biological Chemistry, Biophysics and Bioengineering, School of Engineering and Physical Sciences, Heriot-Watt University, Heriot-Watt Research Park, Edinburgh EH14 4AS, UK
| |
Collapse
|
124
|
Wang Y, Xiong Z, Qiao Y, Zhang Q, Zhou G, Zhou C, Ma X, Jiang X, Yu W. Acetyl-11-keto-beta-boswellic acid modulates macrophage polarization and Schwann cell migration to accelerate spinal cord injury repair in rats. CNS Neurosci Ther 2024; 30:e14642. [PMID: 38430464 PMCID: PMC10908365 DOI: 10.1111/cns.14642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 01/12/2024] [Accepted: 01/28/2024] [Indexed: 03/03/2024] Open
Abstract
BACKGROUND Inhibiting secondary inflammatory damage caused by glial cells and creating a stable microenvironment is one of the main strategies to investigate drugs for the treatment of spinal cord injury. Acetyl-11-keto-beta-boswellic acid (AKBA) is the active component of the natural drug boswellia, which has anti-inflammatory and antioxidant effects and offers a possible therapeutic option for spinal cord injury. METHODS In this study, a spinal cord injury model was established by crushing spinal cord, respectively, to detect the M1 macrophage inflammatory markers: iNOS, TNF-α, IL-1β, and the M2 macrophage markers CD206, ARG-1, IL-10, and the detection of antioxidant enzymes and MDA. In vitro, macrophages were cultured to verify the main mechanism of the macrophage switch from Nrf2/HO-1 to M2 type by flow cytometry, immunofluorescence, and other techniques. Macrophage and Schwann cell co-culture validated the migration mechanism of Schwann cells promoted by AKBA. RESULTS AKBA significantly enhanced the antioxidant enzyme activities of CAT, GSH-Px, T-AOC, and SOD, reduced MDA content, and reduced oxidative damage caused by spinal cord injury via the Nrf2/HO-1 signaling pathway; AKBA mediates Nrf2/HO-1/IL-10, converts macrophages from M1 to M2 type, reduces inflammation, and promotes Schwann cell migration, thereby accelerating the repair of spinal cord injury in rats. CONCLUSIONS Our work demonstrates that AKBA can attenuate oxidative stress as well as the secondary inflammatory injury caused by macrophages after SCI, promote Schwann cell migration to the injury site, and thus accelerate the repair of the injured spinal cord.
Collapse
Affiliation(s)
- Yao Wang
- Department of Veterinary MedicineNortheast Agricultural UniversityHarbinHeilongjiangChina
| | - Zongliang Xiong
- Department of Veterinary MedicineNortheast Agricultural UniversityHarbinHeilongjiangChina
| | - Yuncong Qiao
- School of Life SciencesNortheast Agricultural UniversityHarbinHeilongjiangChina
| | - Qiyuan Zhang
- Department of Veterinary MedicineNortheast Agricultural UniversityHarbinHeilongjiangChina
| | - Guanghu Zhou
- Department of Veterinary MedicineNortheast Agricultural UniversityHarbinHeilongjiangChina
| | - Chong Zhou
- Department of Veterinary MedicineNortheast Agricultural UniversityHarbinHeilongjiangChina
| | - Xianglin Ma
- Department of Veterinary MedicineNortheast Agricultural UniversityHarbinHeilongjiangChina
| | - Xiaowen Jiang
- Department of Veterinary MedicineNortheast Agricultural UniversityHarbinHeilongjiangChina
| | - Wenhui Yu
- Department of Veterinary MedicineNortheast Agricultural UniversityHarbinHeilongjiangChina
- Key Laboratory of the Provincial Education, Department of Heilongjiang for Common Animal Disease Prevention and TreatmentNortheast Agricultural UniversityHarbinHeilongjiangChina
- Institute of Chinese Veterinary MedicineNortheast Agricultural UniversityHarbinHeilongjiangChina
| |
Collapse
|
125
|
Gu J, Zhang P, Li H, Wang Y, Huang Y, Fan L, Ma X, Qian X, Xi J. Cerium-Luteolin Nanocomplexes in Managing Inflammation-Related Diseases by Antioxidant and Immunoregulation. ACS NANO 2024; 18:6229-6242. [PMID: 38345570 DOI: 10.1021/acsnano.3c09528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Abstract
Oxidative stress, characterized by an imbalance between reactive oxygen species (ROS) production and the antioxidant defense system, plays a pivotal role in inflammation-related diseases. Excessive ROS levels can induce cellular damage and impair normal physiological functions, triggering the release of inflammatory mediators and exacerbating the inflammatory response, ultimately leading to irreversible tissue damage. In this study, we synthesized cerium ion-luteolin nanocomplexes (CeLutNCs) by coordinating Ce ions with the natural product luteolin, aiming to develop a therapeutic agent with excellent antioxidant and immunoregulation properties for ROS-related inflammation treatment. In vitro experiments demonstrated that the prepared CeLutNCs effectively scavenged excess ROS, prevented cell apoptosis, down-regulated levels of important inflammatory cytokines, regulated the response of inflammatory macrophages, and suppressed the activation of the nuclear factor-κ-gene binding (NF-κB) pathway. In an acute kidney injury (AKI) animal model, CeLutNCs exhibited significant efficacy in improving kidney function, repairing damaged renal tissue, and reducing oxidative stress, inflammatory response, and cellular apoptosis. Moreover, the therapeutic potential of CeLutNCs in an acute lung injury (ALI) model was confirmed through the assessment of inflammatory responses and histopathological studies. This study emphasizes the effectiveness of these metal-natural product coordination nanocomplexes as a promising therapeutic approach for preventing AKI and other diseases associated with oxidative stress.
Collapse
Affiliation(s)
- Jiake Gu
- Medical College, Institute of Translational Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou, Jiangsu 225009, China
| | - Peiying Zhang
- Medical College, Institute of Translational Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, China
| | - Huajun Li
- Medical College, Institute of Translational Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, China
| | - Yisen Wang
- Medical College, Institute of Translational Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, China
| | - Ying Huang
- Medical College, Institute of Translational Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, China
| | - Lei Fan
- School of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou, Jiangsu 225002, China
| | - Xiao Ma
- Department of Cardiology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Xiaodong Qian
- Department of Cardiology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Juqun Xi
- Medical College, Institute of Translational Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou, Jiangsu 225009, China
| |
Collapse
|
126
|
Shin JJ, Park J, Shin HS, Arab I, Suk K, Lee WH. Roles of lncRNAs in NF-κB-Mediated Macrophage Inflammation and Their Implications in the Pathogenesis of Human Diseases. Int J Mol Sci 2024; 25:2670. [PMID: 38473915 DOI: 10.3390/ijms25052670] [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: 01/31/2024] [Revised: 02/20/2024] [Accepted: 02/22/2024] [Indexed: 03/14/2024] Open
Abstract
Over the past century, molecular biology's focus has transitioned from proteins to DNA, and now to RNA. Once considered merely a genetic information carrier, RNA is now recognized as both a vital element in early cellular life and a regulator in complex organisms. Long noncoding RNAs (lncRNAs), which are over 200 bases long but do not code for proteins, play roles in gene expression regulation and signal transduction by inducing epigenetic changes or interacting with various proteins and RNAs. These interactions exhibit a range of functions in various cell types, including macrophages. Notably, some macrophage lncRNAs influence the activation of NF-κB, a crucial transcription factor governing immune and inflammatory responses. Macrophage NF-κB is instrumental in the progression of various pathological conditions including sepsis, atherosclerosis, cancer, autoimmune disorders, and hypersensitivity. It orchestrates gene expression related to immune responses, inflammation, cell survival, and proliferation. Consequently, its malfunction is a key contributor to the onset and development of these diseases. This review aims to summarize the function of lncRNAs in regulating NF-κB activity in macrophage activation and inflammation, with a particular emphasis on their relevance to human diseases and their potential as therapeutic targets. The insights gained from studies on macrophage lncRNAs, as discussed in this review, could provide valuable knowledge for the development of treatments for various pathological conditions involving macrophages.
Collapse
Affiliation(s)
- Jae-Joon Shin
- School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Jeongkwang Park
- School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Hyeung-Seob Shin
- School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Imene Arab
- School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Kyoungho Suk
- Department of Pharmacology, Brain Science and Engineering Institute, BK21 Plus KNU Biomedical Convergence Program, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Won-Ha Lee
- School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Republic of Korea
| |
Collapse
|
127
|
Song Y, Hu J, Ma C, Liu H, Li Z, Yang Y. Macrophage-Derived Exosomes as Advanced Therapeutics for Inflammation: Current Progress and Future Perspectives. Int J Nanomedicine 2024; 19:1597-1627. [PMID: 38406601 PMCID: PMC10888065 DOI: 10.2147/ijn.s449388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 02/10/2024] [Indexed: 02/27/2024] Open
Abstract
The development of numerous diseases is significantly influenced by inflammation. Macrophage-derived exosomes (M-Exos) play a role in controlling inflammatory reactions in various conditions, including chronic inflammatory pain, hypertension, and diabetes. However, the specific targets and roles of M-Exos in regulating inflammation in diseases remain largely unknown. This review summarizes current knowledge on M-Exos biogenesis and provides updated information on M-Exos' biological function in inflammation modulation. Furthermore, this review highlights the functionalization and engineering strategies of M-Exos, while providing an overview of cutting-edge approaches to engineering M-Exos and advancements in their application as therapeutics for inflammation modulation. Finally, multiple engineering strategies and mechanisms are presented in this review along with their perspectives and challenges, and the potential contribution that M-Exos may have in diseases through the modulation of inflammation is discussed.
Collapse
Affiliation(s)
- Yanjuan Song
- Graduate School, Wuhan Sports University, Wuhan, Hubei Province, People’s Republic of China
| | - Jing Hu
- Wuhan Children’s Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, Hubei Province, People’s Republic of China
| | - Chunlian Ma
- Fitness Monitoring and Chronic Disease Intervention Research Center, Wuhan Sports University, Wuhan, Hubei Province, People’s Republic of China
- College of Sports Medicine, Wuhan Sports University, Wuhan, Hubei Province, People’s Republic of China
- Hubei Key Laboratory of Exercise Training and Monitoring, Wuhan Sports University, Wuhan, Hubei Province, People’s Republic of China
| | - Hua Liu
- Fitness Monitoring and Chronic Disease Intervention Research Center, Wuhan Sports University, Wuhan, Hubei Province, People’s Republic of China
- College of Sports Medicine, Wuhan Sports University, Wuhan, Hubei Province, People’s Republic of China
- Hubei Key Laboratory of Exercise Training and Monitoring, Wuhan Sports University, Wuhan, Hubei Province, People’s Republic of China
| | - Zhanghua Li
- Department of Orthopedics, Wuhan Third Hospital, Tongren Hospital of Wuhan University, Wuhan, Hubei Province, People’s Republic of China
| | - Yi Yang
- Fitness Monitoring and Chronic Disease Intervention Research Center, Wuhan Sports University, Wuhan, Hubei Province, People’s Republic of China
- College of Sports Medicine, Wuhan Sports University, Wuhan, Hubei Province, People’s Republic of China
- Hubei Key Laboratory of Exercise Training and Monitoring, Wuhan Sports University, Wuhan, Hubei Province, People’s Republic of China
| |
Collapse
|
128
|
Salvan da Rosa J, Bramorski Mohr ET, Lubschinski TL, Vieira GN, Rossa TA, Mandolesi Sá M, Dalmarco EM. Interference in Macrophage Balance (M1/M2): The Mechanism of Action Responsible for the Anti-Inflammatory Effect of a Fluorophenyl-Substituted Imidazole. Mediators Inflamm 2024; 2024:9528976. [PMID: 38405621 PMCID: PMC10894048 DOI: 10.1155/2024/9528976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 01/19/2024] [Accepted: 02/03/2024] [Indexed: 02/27/2024] Open
Abstract
Traditionally, the treatment of inflammatory conditions has focused on the inhibition of inflammatory mediator production; however, many conditions are refractory to this classical approach. Recently, an alternative has been presented by researchers to solve this problem: The immunomodulation of cells closely related to inflammation. Hence, macrophages, a critical key in both innate and acquired immunity, have been presented as an alternative target for the development of new medicines. In this work, we tested the fluorophenyl-imidazole for its anti-inflammatory activity and possible immunomodulatory effect on RAW 264.7 macrophages. We also evaluated the anti-inflammatory effect of the compound, and the macrophage repolarization to M2 was confirmed by the ability of the compound to reduce the M1 markers TNF-α, IL-6, MCP-1, IL-12p70, IFN-γ, and TLR4, the high levels of p65 phosphorylated, iNOS and COX-2 mRNA expression, and the fact that the compound was not able to induce the production of M1 markers when used in macrophages without lipopolysaccharide (LPS) stimulation. Moreover, fluorophenyl-imidazole had the ability to increase the M2 markers IL-4, IL-13, CD206, apoptosis and phagocytosis levels, arginase-1, and FIZZ-1 mRNA expression before LPS stimulation. Similarly, it was also able to induce the production of these same M2 markers in macrophages without being induced with LPS. These results reinforce the affirmation that the fluorophenyl-imidazole has an important anti-inflammatory effect and demonstrates that this effect is due to immunomodulatory activity, having the ability to trigger a repolarization of macrophages from M1 to M2a. These facts suggest that this molecule could be used as an alternative scaffold for the development of a new medicine to treat inflammatory conditions, where the anti-inflammatory and proregenerative properties of M2a macrophages are desired.
Collapse
Affiliation(s)
- Julia Salvan da Rosa
- Department of Clinical Analysis, Center for Health Sciences, Campus Universitário—Trindade, Universidade Federal de Santa Catarina, Florianópolis 88040-970, SC, Brazil
| | - Eduarda Talita Bramorski Mohr
- Department of Clinical Analysis, Center for Health Sciences, Campus Universitário—Trindade, Universidade Federal de Santa Catarina, Florianópolis 88040-970, SC, Brazil
| | - Tainá Larissa Lubschinski
- Department of Clinical Analysis, Center for Health Sciences, Campus Universitário—Trindade, Universidade Federal de Santa Catarina, Florianópolis 88040-970, SC, Brazil
| | - Guilherme Nicácio Vieira
- Department of Clinical Analysis, Center for Health Sciences, Campus Universitário—Trindade, Universidade Federal de Santa Catarina, Florianópolis 88040-970, SC, Brazil
| | - Thais Andreia Rossa
- Department of Chemistry, Center for Physical and Mathematical Sciences, Campus Universitário—Trindade, Universidade Federal de Santa Catarina, Florianópolis 88040-970, SC, Brazil
| | - Marcus Mandolesi Sá
- Department of Chemistry, Center for Physical and Mathematical Sciences, Campus Universitário—Trindade, Universidade Federal de Santa Catarina, Florianópolis 88040-970, SC, Brazil
| | - Eduardo Monguilhott Dalmarco
- Department of Clinical Analysis, Center for Health Sciences, Campus Universitário—Trindade, Universidade Federal de Santa Catarina, Florianópolis 88040-970, SC, Brazil
| |
Collapse
|
129
|
Zhao C, Luo Q, Huang J, Su S, Zhang L, Zheng D, Chen M, Lin X, Zhong J, Li L, Ling K, Zhang S. Extracellular Vesicles Derived from Human Adipose-Derived Mesenchymal Stem Cells Alleviate Sepsis-Induced Acute Lung Injury through a MicroRNA-150-5p-Dependent Mechanism. ACS Biomater Sci Eng 2024; 10:946-959. [PMID: 38154081 DOI: 10.1021/acsbiomaterials.3c00614] [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] [Indexed: 12/30/2023]
Abstract
Extracellular vesicles (EVs) derived from human adipose mesenchymal stem cells (hADSCs) may exert a therapeutic benefit in alleviating sepsis-induced organ dysfunction by delivering cargos that include RNAs and proteins to target cells. The current study aims to explore the protective effect of miR-150-5p delivered by hADSC-EVs on sepsis-induced acute lung injury (ALI). We noted low expression of miR-150-5p in plasma and bronchoalveolar lavage fluid samples from patients with sepsis-induced ALI. The hADSC-EVs were isolated and subsequently cocultured with macrophages. It was established that hADSC-EVs transferred miR-150-5p to macrophages, where miR-150-5p targeted HMGA2 to inhibit its expression and, consequently, inactivated the MAPK pathway. This effect contributed to the promotion of M2 polarization of macrophages and the inhibition of proinflammatory cytokines. Further, mice were made septic by cecal ligation and puncture in vivo and treated with hADSC-EVs to elucidate the effect of hADSC-EVs on sepsis-induced ALI. The in vivo experimental results confirmed a suppressive role of hADSC-EVs in sepsis-induced ALI. Our findings suggest that hADSC-EV-mediated transfer of miR-150-5p may be a novel mechanism underlying the paracrine effects of hADSC-EVs on the M2 polarization of macrophages in sepsis-induced ALI.
Collapse
Affiliation(s)
- Chengkuan Zhao
- Department of Pharmacy, Guangzhou Red Cross Hospital, Jinan University, Guangzhou 510220, P.R. China
| | - Qianhua Luo
- Department of Pharmacology, Guangdong Second Provincial General Hospital, Guangzhou 510317, P.R. China
- Jinshazhou Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510168, P.R. China
| | - Jianxiang Huang
- College of Pharmacy, Jinan University, Guangzhou 510220, P.R. China
| | - Siman Su
- Department of Pharmacology, Shantou University Medical College, Shantou 515041, P.R. China
| | - Lijuan Zhang
- Department of Pharmacy, YueBei People's Hospital (YueBei People's Hospital affiliated to Shantou University Medical College), ShaoGuan 512000, P.R. China
| | - Danling Zheng
- Department of Pharmacy, Guangzhou Red Cross Hospital, Jinan University, Guangzhou 510220, P.R. China
- Department of Pharmacology, Shantou University Medical College, Shantou 515041, P.R. China
| | - Meini Chen
- Department of Pharmacology, Shantou University Medical College, Shantou 515041, P.R. China
| | - Xinyue Lin
- Department of Pharmacology, Shantou University Medical College, Shantou 515041, P.R. China
| | - Jialin Zhong
- Department of Pharmacy, Guangzhou Red Cross Hospital, Jinan University, Guangzhou 510220, P.R. China
| | - Li Li
- Department of Pharmacy, Guangzhou Red Cross Hospital, Jinan University, Guangzhou 510220, P.R. China
| | - Kai Ling
- Department of Pharmacology, Shantou University Medical College, Shantou 515041, P.R. China
| | - Shuyao Zhang
- Department of Pharmacy, Guangzhou Red Cross Hospital, Jinan University, Guangzhou 510220, P.R. China
| |
Collapse
|
130
|
Zhang X, Deng C, Qi S. Periosteum Containing Implicit Stem Cells: A Progressive Source of Inspiration for Bone Tissue Regeneration. Int J Mol Sci 2024; 25:2162. [PMID: 38396834 PMCID: PMC10889827 DOI: 10.3390/ijms25042162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 01/12/2024] [Accepted: 01/26/2024] [Indexed: 02/25/2024] Open
Abstract
The periosteum is known as the thin connective tissue covering most bone surfaces. Its extrusive bone regeneration capacity was confirmed from the very first century-old studies. Recently, pluripotent stem cells in the periosteum with unique physiological properties were unveiled. Existing in dynamic contexts and regulated by complex molecular networks, periosteal stem cells emerge as having strong capabilities of proliferation and multipotential differentiation. Through continuous exploration of studies, we are now starting to acquire more insight into the great potential of the periosteum in bone formation and repair in situ or ectopically. It is undeniable that the periosteum is developing further into a more promising strategy to be harnessed in bone tissue regeneration. Here, we summarized the development and structure of the periosteum, cell markers, and the biological features of periosteal stem cells. Then, we reviewed their pivotal role in bone repair and the underlying molecular regulation. The understanding of periosteum-related cellular and molecular content will help enhance future research efforts and application transformation of the periosteum.
Collapse
Affiliation(s)
- Xinyuan Zhang
- Department of Prosthodontics, Shanghai Stomatological Hospital, School of Stomatology, Fudan University, Shanghai 200001, China;
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai 200001, China
| | - Chen Deng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China;
| | - Shengcai Qi
- Department of Prosthodontics, Shanghai Stomatological Hospital, School of Stomatology, Fudan University, Shanghai 200001, China;
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai 200001, China
| |
Collapse
|
131
|
Chen Y, Markov N, Gigon L, Hosseini A, Yousefi S, Stojkov D, Simon HU. The BK Channel Limits the Pro-Inflammatory Activity of Macrophages. Cells 2024; 13:322. [PMID: 38391935 PMCID: PMC10886595 DOI: 10.3390/cells13040322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 01/31/2024] [Accepted: 02/07/2024] [Indexed: 02/24/2024] Open
Abstract
Macrophages play a crucial role in the innate immune response, serving as key effector cells in the defense against pathogens. Although the role of the large-conductance voltage and calcium-activated potassium channel, also known as the KCa1.1 or BK channel, in regulating neurotransmitter release and smooth muscle contraction is well known, its potential involvement in immune regulation remains unclear. We employed BK-knockout macrophages and noted that the absence of a BK channel promotes the polarization of macrophages towards a pro-inflammatory phenotype known as M1 macrophages. Specifically, the absence of the BK channel resulted in a significant increase in the secretion of the pro-inflammatory cytokine IL-6 and enhanced the activity of extracellular signal-regulated kinases 1 and 2 (Erk1/2 kinases), Ca2+/calmodulin-dependent protein kinase II (CaMKII), and the transcription factor ATF-1 within M1 macrophages. Additionally, the lack of the BK channel promoted the activation of the AIM2 inflammasome without affecting the activation of the NLRC4 and NLRP3 inflammasomes. To further investigate the role of the BK channel in regulating AIM2 inflammasome activation, we utilized BK channel inhibitors, such as paxilline and iberiotoxin, along with the BK channel activator NS-11021. Pharmacological inactivation of the BK channel increased, and its stimulation inhibited IL-1β production following AIM2 inflammasome activation in wild-type macrophages. Moreover, wild-type macrophages displayed increased calcium influx when activated with the AIM2 inflammasome, whereas BK-knockout macrophages did not due to the impaired extracellular calcium influx upon activation. Furthermore, under conditions of a calcium-free medium, IL-1β production following AIM2 inflammasome activation was increased in both wild-type and BK-knockout macrophages. This suggests that the BK channel is required for the influx of extracellular calcium in macrophages, thus limiting AIM2 inflammasome activation. In summary, our study reveals a regulatory role of the BK channel in macrophages under inflammatory conditions.
Collapse
Affiliation(s)
- Yihe Chen
- Institute of Pharmacology, University of Bern, 3010 Bern, Switzerland
| | - Nikita Markov
- Institute of Pharmacology, University of Bern, 3010 Bern, Switzerland
| | - Lea Gigon
- Institute of Pharmacology, University of Bern, 3010 Bern, Switzerland
| | - Aref Hosseini
- Institute of Pharmacology, University of Bern, 3010 Bern, Switzerland
| | - Shida Yousefi
- Institute of Pharmacology, University of Bern, 3010 Bern, Switzerland
| | - Darko Stojkov
- Institute of Pharmacology, University of Bern, 3010 Bern, Switzerland
| | - Hans-Uwe Simon
- Institute of Pharmacology, University of Bern, 3010 Bern, Switzerland
- Institute of Biochemistry, Brandenburg Medical School, 16816 Neuruppin, Germany
| |
Collapse
|
132
|
Shen Y, Xu Y, Yu Z, Chen G, Chen B, Liao L. Multifunctional Injectable Microspheres Containing "Naturally-Derived" Photothermal Transducer for Synergistic Physical and Chemical Treating of Acute Osteomyelitis through Sequential Immunomodulation. ACS NANO 2024. [PMID: 38335113 DOI: 10.1021/acsnano.3c10697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/12/2024]
Abstract
Osteomyelitis induced by Staphylococcus aureus (S. aureus) is a persistent and deep-seated infection that affects bone tissue. The main challenges in treating osteomyelitis include antibiotic resistance, systemic toxicity, and the need for multiple recurrent surgeries. An ideal therapeutic strategy involves the development of materials that combine physical, chemical, and immunomodulatory synergistic effects. In this work, we prepared injectable microspheres consisting of an interpenetrating network of ionic-cross-linked sodium alginate (SA) and genipin (Gp)-cross-linked gelatin (Gel) incorporated with tannic acid (TA) and copper ions (Cu2+). The Gp-cross-linked Gel acted as a "naturally-derived" photothermal therapy (PTT) agent. The results showed that the microspheres exhibited efficient and rapid bactericidal effects against both S. aureus and Escherichia coli (E. coli) under the irradiation of near-infrared light at 808 nm wavelength; moreover, the release of Cu2+ also induced sustained inhibitory effects against bacteria during the nonirradiation period. The in vitro cell culture results indicated that when combined with PTT, the microspheres could adaptively modulate macrophage M1 and M2 phenotypes in sequence. Additionally, these microspheres were found to enhance the osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs). In vivo studies conducted in a rat femur osteomyelitis model with bone defects showed that under multiple laser irradiation the microspheres effectively controlled bacterial infection, improved the pathological immune microenvironment, and significantly enhanced the repair and regeneration of bone tissues in the affected area.
Collapse
Affiliation(s)
- Yang Shen
- Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou 510515, China
| | - Yaowen Xu
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Ziqian Yu
- Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou 510515, China
| | - Guo Chen
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Bin Chen
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Liqiong Liao
- Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
133
|
O'Connell P. Uncovering mechanisms underlying complement-mediated cancer immune evasion. Mol Ther 2024; 32:277-278. [PMID: 38246164 PMCID: PMC10862000 DOI: 10.1016/j.ymthe.2024.01.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 01/10/2024] [Accepted: 01/10/2024] [Indexed: 01/23/2024] Open
Affiliation(s)
- Patrick O'Connell
- Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Genetics and Genomic Sciences, The Mount Sinai Hospital, New York, NY, USA; Department of Pediatrics, The Mount Sinai Hospital, New York, NY, USA.
| |
Collapse
|
134
|
Kesapragada M, Sun YH, Zlobina K, Recendez C, Fregoso D, Yang HY, Aslankoohi E, Isseroff R, Rolandi M, Zhao M, Gomez M. Deep learning classification for macrophage subtypes through cell migratory pattern analysis. Front Cell Dev Biol 2024; 12:1259037. [PMID: 38385029 PMCID: PMC10879298 DOI: 10.3389/fcell.2024.1259037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Accepted: 01/22/2024] [Indexed: 02/23/2024] Open
Abstract
Macrophages can exhibit pro-inflammatory or pro-reparatory functions, contingent upon their specific activation state. This dynamic behavior empowers macrophages to engage in immune reactions and contribute to tissue homeostasis. Understanding the intricate interplay between macrophage motility and activation status provides valuable insights into the complex mechanisms that govern their diverse functions. In a recent study, we developed a classification method based on morphology, which demonstrated that movement characteristics, including speed and displacement, can serve as distinguishing factors for macrophage subtypes. In this study, we develop a deep learning model to explore the potential of classifying macrophage subtypes based solely on raw trajectory patterns. The classification model relies on the time series of x-y coordinates, as well as the distance traveled and net displacement. We begin by investigating the migratory patterns of macrophages to gain a deeper understanding of their behavior. Although this analysis does not directly inform the deep learning model, it serves to highlight the intricate and distinct dynamics exhibited by different macrophage subtypes, which cannot be easily captured by a finite set of motility metrics. Our study uses cell trajectories to classify three macrophage subtypes: M0, M1, and M2. This advancement holds promising implications for the future, as it suggests the possibility of identifying macrophage subtypes without relying on shape analysis. Consequently, it could potentially eliminate the necessity for high-quality imaging techniques and provide more robust methods for analyzing inherently blurry images.
Collapse
Affiliation(s)
- Manasa Kesapragada
- Department of Applied Mathematics, Baskin School of Engineering, University of California, Santa Cruz, Santa Cruz, CA, United States
| | - Yao-Hui Sun
- Department of Ophthalmology and Vision Science, School of Medicine, University of California, Davis, Sacramento, CA, United States
| | - Ksenia Zlobina
- Department of Applied Mathematics, Baskin School of Engineering, University of California, Santa Cruz, Santa Cruz, CA, United States
| | - Cynthia Recendez
- Department of Ophthalmology and Vision Science, School of Medicine, University of California, Davis, Sacramento, CA, United States
| | - Daniel Fregoso
- Department of Dermatology, School of Medicine, UC Davis, Sacramento, CA, United States
| | - Hsin-Ya Yang
- Department of Dermatology, School of Medicine, UC Davis, Sacramento, CA, United States
| | - Elham Aslankoohi
- Department of Electrical and Computer Engineering, Baskin School of Engineering, University of California, Santa Cruz, Santa Cruz, CA, United States
| | - Rivkah Isseroff
- Department of Dermatology, School of Medicine, UC Davis, Sacramento, CA, United States
| | - Marco Rolandi
- Department of Electrical and Computer Engineering, Baskin School of Engineering, University of California, Santa Cruz, Santa Cruz, CA, United States
| | - Min Zhao
- Department of Ophthalmology and Vision Science, School of Medicine, University of California, Davis, Sacramento, CA, United States
- Department of Dermatology, School of Medicine, UC Davis, Sacramento, CA, United States
| | - Marcella Gomez
- Department of Applied Mathematics, Baskin School of Engineering, University of California, Santa Cruz, Santa Cruz, CA, United States
| |
Collapse
|
135
|
Ruelas Castillo J, Neupane P, Karanika S, Krug S, Quijada D, Garcia A, Ayeh S, Yilma A, Costa DL, Sher A, Fotouhi N, Serbina N, Karakousis PC. The heme oxygenase-1 metalloporphyrin inhibitor stannsoporfin enhances the bactericidal activity of a novel regimen for multidrug-resistant tuberculosis in a murine model. Antimicrob Agents Chemother 2024; 68:e0104323. [PMID: 38132181 PMCID: PMC10848751 DOI: 10.1128/aac.01043-23] [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: 08/10/2023] [Accepted: 11/17/2023] [Indexed: 12/23/2023] Open
Abstract
Multidrug-resistant (MDR) Mycobacterium tuberculosis (Mtb) poses significant challenges to global tuberculosis (TB) control efforts. Host-directed therapies (HDTs) offer a novel approach to TB treatment by enhancing immune-mediated clearance of Mtb. Prior preclinical studies found that the inhibition of heme oxygenase-1 (HO-1), an enzyme involved in heme metabolism, with tin-protoporphyrin IX (SnPP) significantly reduced mouse lung bacillary burden when co-administered with the first-line antitubercular regimen. Here, we evaluated the adjunctive HDT activity of a novel HO-1 inhibitor, stannsoporfin (SnMP), in combination with a novel MDR-TB regimen comprising a next-generation diarylquinoline, TBAJ-876 (S), pretomanid (Pa), and a new oxazolidinone, TBI-223 (O) (collectively, SPaO), in Mtb-infected BALB/c mice. After 4 weeks of treatment, SPaO + SnMP 5mg/kg reduced mean lung bacillary burden by an additional 0.69 log10 (P = 0.01) relative to SPaO alone. As early as 2 weeks post-treatment initiation, SnMP adjunctive therapy differentially altered the expression of pro-inflammatory cytokine genes and CD38, a marker of M1 macrophages. Next, we evaluated the sterilizing potential of SnMP adjunctive therapy in a mouse model of microbiological relapse. After 6 weeks of treatment, SPaO + SnMP 10mg/kg reduced lung bacterial burdens to 0.71 ± 0.23 log10 colony-forming units (CFUs), a 0.78 log-fold greater decrease in lung CFU compared to SpaO alone (P = 0.005). However, adjunctive SnMP did not reduce microbiological relapse rates after 5 or 6 weeks of treatment. SnMP was well tolerated and did not significantly alter gross or histological lung pathology. SnMP is a promising HDT candidate requiring further study in combination with regimens for drug-resistant TB.
Collapse
Affiliation(s)
- Jennie Ruelas Castillo
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Pranita Neupane
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Styliani Karanika
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Stefanie Krug
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Darla Quijada
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Andrew Garcia
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Samuel Ayeh
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Addis Yilma
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Diego L. Costa
- Departmento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Alan Sher
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | | | | | - Petros C. Karakousis
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| |
Collapse
|
136
|
Luan X, Lei T, Fang J, Liu X, Fu H, Li Y, Chu W, Jiang P, Tong C, Qi H, Fu Y. Blockade of C5a receptor unleashes tumor-associated macrophage antitumor response and enhances CXCL9-dependent CD8 + T cell activity. Mol Ther 2024; 32:469-489. [PMID: 38098230 PMCID: PMC10861991 DOI: 10.1016/j.ymthe.2023.12.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 09/17/2023] [Accepted: 12/11/2023] [Indexed: 12/25/2023] Open
Abstract
Macrophages play a crucial role in shaping the immune state within the tumor microenvironment (TME) and are often influenced by tumors to hinder antitumor immunity. However, the underlying mechanisms are still elusive. Here, we observed abnormal expression of complement 5a receptor (C5aR) in human ovarian cancer (OC), and identified high levels of C5aR expression on tumor-associated macrophages (TAMs), which led to the polarization of TAMs toward an immunosuppressive phenotype. C5aR knockout or inhibitor treatment restored TAM antitumor response and attenuated tumor progression. Mechanistically, C5aR deficiency reprogrammed macrophages from a protumor state to an antitumor state, associating with the upregulation of immune response and stimulation pathways, which in turn resulted in the enhanced antitumor response of cytotoxic T cells in a manner dependent on chemokine (C-X-C motif) ligand 9 (CXCL9). The pharmacological inhibition of C5aR also improved the efficacy of immune checkpoint blockade therapy. In patients, C5aR expression associated with CXCL9 production and infiltration of CD8+ T cells, and a high C5aR level predicted poor clinical outcomes and worse benefits from anti-PD-1 therapy. Thus, our study sheds light on the mechanisms underlying the modulation of TAM antitumor immune response by the C5a-C5aR axis and highlights the potential of targeting C5aR for clinical applications.
Collapse
Affiliation(s)
- Xiaojin Luan
- Chongqing Key Laboratory of Maternal and Fetal Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Ting Lei
- Chongqing Key Laboratory of Maternal and Fetal Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Jie Fang
- Department of Gynecology, The Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang 212001, Jiangsu, China
| | - Xue Liu
- Chongqing Key Laboratory of Maternal and Fetal Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Obstetrics, Yongchuan Hospital of Chongqing Medical University, Chongqing 402160, China
| | - Huijia Fu
- Department of Reproductive Medicine Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Yiran Li
- Chongqing Key Laboratory of Maternal and Fetal Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Wei Chu
- Chongqing Key Laboratory of Maternal and Fetal Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Peng Jiang
- Department of Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Chao Tong
- Chongqing Key Laboratory of Maternal and Fetal Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.
| | - Hongbo Qi
- Chongqing Key Laboratory of Maternal and Fetal Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Women and Children's Hospital of Chongqing Medical University, Chongqing 401147, China.
| | - Yong Fu
- Chongqing Key Laboratory of Maternal and Fetal Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
137
|
Xiang Y, Wang G, Liu B, Zheng H, Liu Q, Ma G, Du J. Macrophage-Related Gene Signatures for Predicting Prognosis and Immunotherapy of Lung Adenocarcinoma by Machine Learning and Bioinformatics. J Inflamm Res 2024; 17:737-754. [PMID: 38348277 PMCID: PMC10859764 DOI: 10.2147/jir.s443240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 01/30/2024] [Indexed: 02/15/2024] Open
Abstract
Background In recent years, the immunotherapy of lung adenocarcinoma has developed rapidly, but the good therapeutic effect only exists in some patients, and most of the current predictors cannot predict it very well. Tumor-infiltrating macrophages have been reported to play a crucial role in lung adenocarcinoma (LUAD). Thus, we want to build novel molecular markers based on macrophages. Methods By non-negative matrix factorization (NMF) algorithm and Cox regression analysis, we constructed macrophage-related subtypes of LUAD patients and built a novel gene signature consisting of 12 differentially expressed genes between two subtypes. The gene signature was further validated in Gene-Expression Omnibus (GEO) datasets. Its predictive effect on prognosis and immunotherapy outcome was further evaluated with rounded analyses. We finally explore the role of TRIM28 in LUAD with a series of in vitro experiments. Results Our research indicated that a higher LMS score was significantly correlated with tumor staging, pathological grade, tumor node metastasis stage, and survival. LMS was identified as an independent risk factor for OS in LUAD patients and verified in GEO datasets. Clinical response to immunotherapy was better in patients with low LMS score compared to those with high LMS score. TRIM28, a key gene in the gene signature, was shown to promote the proliferation, invasion and migration of LUAD cell. Conclusion Our study highlights the significant role of gene signature in predicting the prognosis and immunotherapy efficacy of LUAD patients, and identifies TRIM28 as a potential biomarker for the treatment of LUAD.
Collapse
Affiliation(s)
- Yunzhi Xiang
- Institute of Oncology, Shandong Provincial Hospital, Shandong University, Jinan, People’s Republic of China
| | - Guanghui Wang
- Institute of Oncology, Shandong Provincial Hospital, Shandong University, Jinan, People’s Republic of China
| | - Baoliang Liu
- Institute of Oncology, Shandong Provincial Hospital, Shandong University, Jinan, People’s Republic of China
| | - Haotian Zheng
- Institute of Oncology, Shandong Provincial Hospital, Shandong University, Jinan, People’s Republic of China
| | - Qiang Liu
- Institute of Oncology, Shandong Provincial Hospital, Shandong University, Jinan, People’s Republic of China
| | - Guoyuan Ma
- Institute of Oncology, Shandong Provincial Hospital, Shandong University, Jinan, People’s Republic of China
| | - Jiajun Du
- Institute of Oncology, Shandong Provincial Hospital, Shandong University, Jinan, People’s Republic of China
| |
Collapse
|
138
|
Liang J, Chen Y, Wang Z, Wang Y, Mu S, Zhang D, Wang Z, Zeng W. Exploring the association between rosacea and acne by integrated bioinformatics analysis. Sci Rep 2024; 14:3065. [PMID: 38321132 PMCID: PMC10847114 DOI: 10.1038/s41598-024-53453-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: 07/04/2023] [Accepted: 01/31/2024] [Indexed: 02/08/2024] Open
Abstract
Clinically, rosacea occurs frequently in acne patients, which hints the existence of shared signals. However, the connection between the pathophysiology of rosacea and acne are not yet fully understood. This study aims to unveil molecular mechanism in the pathogenesis of rosacea and acne. We identified differentially expressed genes (DEGs) by limma and weighted gene co-expression network analysis and screened hub genes by constructing a protein-protein interaction network. The hub genes were verified in different datasets. Then, we performed a correlation analysis between the hub genes and the pathways. Finally, we predicted and verified transcription factors of hub genes, performed the immune cell infiltration analysis using CIBERSORT, and calculated the correlation between hub genes and immune cells. A total of 169 common DEGs were identified, which were mainly enriched in immune-related pathways. Finally, hub genes were identified as IL1B, PTPRC, CXCL8, MMP9, CCL4, CXCL10, CD163, CCR5, CXCR4, and TLR8. 9 transcription factors that regulated the expression of hub genes were identified. The infiltration of γδT cells was significantly increased in rosacea and acne lesions and positively linked with almost all hub genes. These identified hub genes and immune cells may play a crucial role in the development of rosacea and acne.
Collapse
Affiliation(s)
- Jingchen Liang
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Ying Chen
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Zihao Wang
- Department of Cardiology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yawen Wang
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Shengzhi Mu
- Department of Burn and Plastic Surgery, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Dewu Zhang
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Zhao Wang
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| | - Weihui Zeng
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
139
|
Yang L, Tao W, Xie C, Chen Q, Zhao Y, Zhang L, Xiao X, Wang S, Zheng X. Interleukin-37 ameliorates periodontitis development by inhibiting NLRP3 inflammasome activation and modulating M1/M2 macrophage polarization. J Periodontal Res 2024; 59:128-139. [PMID: 37947055 DOI: 10.1111/jre.13196] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 09/04/2023] [Accepted: 10/12/2023] [Indexed: 11/12/2023]
Abstract
OBJECTIVE Our study was designed to explore the role of IL-37 in M1/M2 macrophage polarization imbalance in the pathogenesis of periodontitis. BACKGROUND Periodontitis is a chronic progressive inflammatory disease featured by gingival inflammation and alveolar bone resorption. Recent research has revealed that regulating macrophage polarization is a viable method to ameliorate periodontal inflammation. IL-37 is an anti-inflammatory cytokine, which has been reported to inhibit innate and adaptive immunity. METHODS For in vitro experiment, mouse macrophage RAW264.7 cells were pretreated with 0.1 ng/mL recombinant human IL-37. M1 and M2 polarizations of RAW264.7 cells were induced by 100 ng/mL LPS and 20 ng/mL IL-4, respectively. The expression of M1 (iNOS, TNF-α, and IL-6) and M2 (CD206, Arg1, and IL-10) phenotype markers in RAW264.7 cells was detected by RT-qPCR, western blotting, and immunofluorescence staining. For in vivo experiment, experimental periodontitis mouse models were established by sterile silk ligation (5-0) around the bilateral maxillary second molar of mice for 1 week. H&E staining of the maxillary alveolar bone was used to show the resorption of root cementum and dentin. Alveolar bone loss in mouse models was evaluated through micro-CT analysis. The expression of iNOS and CD206 in gingival tissues was assessed by immunohistochemistry staining. NLRP3 inflammasome activation was confirmed by western blotting. RESULTS IL-37 pretreatment reduced iNOS, TNF-α, and IL-6 expression in LPS-treated RAW264.7 cells but increased CD206, Arg1, and IL-10 in IL-4-treated RAW264.7 cells. LPS-induced upregulation in NLRP3, GSDMD, cleaved-IL-1β, and cleaved-caspase-1 expression was antagonized by IL-37 treatment. In addition, IL-37 administration ameliorated the resorption of root cementum and dentin in periodontitis mouse models. IL-37 prominently decreased iNOS+ cell population but increased CD206+ cell population in gingival tissues of periodontitis mice. The enhancement in NLRP3, GSDMD, cleaved-IL-1β, and cleaved-caspase-1 expression in the gingival tissues of periodontitis mice was offset by IL-37 administration. CONCLUSION IL-37 prevents the progression of periodontitis by suppressing NLRP3 inflammasome activation and mediating M1/M2 macrophage polarization.
Collapse
Affiliation(s)
- Liyan Yang
- Department of Stomatology, The First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Wei Tao
- Department of Stomatology, The First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Chen Xie
- School of Stomatology, Hainan Medical University, Haikou, China
| | - Qiuye Chen
- Department of Stomatology, Hainan Cancer Hospital, Haikou, China
| | - Yunshan Zhao
- Integrated Department, Hainan Stomatological Hospital, Haikou, China
| | - Li Zhang
- School of Stomatology, Hainan Medical University, Haikou, China
| | - Xu Xiao
- Department of Stomatology, The First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Shilu Wang
- Department of Anesthesiology, The First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Xu Zheng
- Department of Stomatology, The First Affiliated Hospital of Hainan Medical University, Haikou, China
- School of Stomatology, Hainan Medical University, Haikou, China
| |
Collapse
|
140
|
Gemmete JJ. Partial Cryoablation and Matrix Metalloproteinase Inhibition May Be a Possible Treatment Option for Hepatocellular Carcinoma. Radiology 2024; 310:e240015. [PMID: 38349243 DOI: 10.1148/radiol.240015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/15/2024]
Affiliation(s)
- Joseph J Gemmete
- From the Department of Radiology, University of Michigan Hospitals, 1500 E Medical Center Dr, UH B1D 328, Ann Arbor, MI 48109
| |
Collapse
|
141
|
Wang H, Yang J, Li X, Zhao H. Current state of immune checkpoints therapy for glioblastoma. Heliyon 2024; 10:e24729. [PMID: 38298707 PMCID: PMC10828821 DOI: 10.1016/j.heliyon.2024.e24729] [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: 10/18/2023] [Revised: 01/12/2024] [Accepted: 01/12/2024] [Indexed: 02/02/2024] Open
Abstract
Glioblastoma (GBM), one of the most aggressive forms of brain cancer, has limited treatment options. Recent years have witnessed the remarkable success of checkpoint inhibitor immunotherapy across various cancer types. Against this backdrop, several clinical trials investigating checkpoint inhibitors for GBM are underway in multiple countries. Furthermore, the integration of immunotherapy with traditional treatment approaches is now emerging as a highly promising strategy. This review summarizes the latest advancements in checkpoint inhibitor immunotherapy for GBM treatment. We provide a concise yet comprehensive overview of current GBM immunotherapy options. Additionally, this review underscores combination strategies and potential biomarkers for predicting response and resistance in GBM immunotherapies.
Collapse
Affiliation(s)
- He Wang
- Department of Neurosurgery, the Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, Shandong, 266005, China
| | - Jing Yang
- Department of Emergency Surgery, the Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, Shandong, 266005, China
| | - Xiangjun Li
- School of medicine, Department of Breast surgery, the Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, Shandong, 266000, China
| | - Hai Zhao
- Department of Neurosurgery, the Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, Shandong, 266005, China
| |
Collapse
|
142
|
Lv X, He Z, Yang M, Wang L, Fu S. Analysis of subsets and localization of macrophages in skin lesions and peripheral blood of patients with keloids. Heliyon 2024; 10:e24034. [PMID: 38283250 PMCID: PMC10818209 DOI: 10.1016/j.heliyon.2024.e24034] [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: 05/29/2023] [Revised: 12/19/2023] [Accepted: 01/02/2024] [Indexed: 01/30/2024] Open
Abstract
Keloids are a type of abnormal fibrous proliferation disease of the skin, characterized by local inflammation that lacks clear pathogenesis and satisfactory treatment. The phenomenon of distinct phenotypes, including M1 and M2 macrophages, is called macrophage polarization. Recently, macrophage polarization has been suggested to play a role in keloid formation. This study aimed to evaluate the relation between macrophage polarization and keloids and identify novel effective treatments for keloids. Differentially expressed genes were identified via RNA sequencing of the skin tissue of healthy controls and patients with keloids and validated using quantitative PCR. Multiplex immunofluorescence microscopy was used to detect different phenotypes of macrophages in keloid tissues. Finally, quantitative PCR validation of differentially expressed genes and flow cytometry were used to analyze macrophages in the peripheral blood of healthy controls and patients with keloids. Total and M2 macrophages were significantly increased in the local skin tissue and peripheral blood of patients with keloids compared with healthy controls. In addition, inflammation- and macrophage polarization-related differentially expressed genes in keloid tissue showed similar expression patterns in the peripheral blood. This study highlighted an increased frequency of total macrophages and M2 polarization in the local skin tissue and peripheral blood of patients with keloids. This systematic macrophage polarization tendency also indicates a potential genetic predisposition to keloids. These findings suggest the possibility of developing new diagnostic and therapeutic indicators for keloids focusing on macrophages.
Collapse
Affiliation(s)
- Xinyi Lv
- Department of Dermatology, The Second Xiangya Hospital of Central South University, Hunan Key Laboratory of Medical Epigenomics, 139 Middle Renmin Road, Changsha, Hunan 410011, China
| | - Zhenghao He
- Department of Dermatology, The Second Xiangya Hospital of Central South University, Hunan Key Laboratory of Medical Epigenomics, 139 Middle Renmin Road, Changsha, Hunan 410011, China
- Department of Plastic Surgery, Zhongshan City People's Hospital, Zhongshan, Guangdong, China
| | - Ming Yang
- Department of Dermatology, The Second Xiangya Hospital of Central South University, Hunan Key Laboratory of Medical Epigenomics, 139 Middle Renmin Road, Changsha, Hunan 410011, China
| | - Lu Wang
- Department of Dermatology, The Second Xiangya Hospital of Central South University, Hunan Key Laboratory of Medical Epigenomics, 139 Middle Renmin Road, Changsha, Hunan 410011, China
| | - Siqi Fu
- Department of Dermatology, The Second Xiangya Hospital of Central South University, Hunan Key Laboratory of Medical Epigenomics, 139 Middle Renmin Road, Changsha, Hunan 410011, China
| |
Collapse
|
143
|
Huang Y, Zhao C, Zheng G, Yuan Y, Gong L, Liu R, An J. Dictamnine Ameliorates DNFB-Induced Atopic Dermatitis Like Skin Lesions in Mice by Inhibiting M1 Macrophage Polarization and Promoting Autophagy. Biol Pharm Bull 2024; 47:175-186. [PMID: 38092386 DOI: 10.1248/bpb.b23-00436] [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] [Indexed: 01/16/2024]
Abstract
Autophagy and M1 macrophage polarization play important roles in the regulation of inflammation in atopic dermatitis (AD). Dictamnine is one of the main ingredients in Cortex Dictamni, a widely used traditional Chinese medicine for the treatment of dermatitis. In the present study, we investigated the anti-inflammatory effects of dictamnine on AD like skin lesions and M1 macrophage polarization. A 2,4-dinitrofluorobenzene (DNFB) triggered AD like skin lesions models in mice was established to identify the ameliorative effects of dictamnine on AD in vivo. In addition, an M1 macrophage polarization model was co-stimulated by lipopolysaccharide (LPS) and interferon-γ (IFN-γ) using phorbol myristate acetate (PMA) differentiated THP-1 cells, to investigate the effect of dictamnine on promoting autophagy and inhibiting inflammatory factor release. Dictamnine suppressed DNFB-induced skin inflammation by inhibiting M1 macrophage polarization, up-regulating the expression of microtubule-associated protein 1A/1B-light chain 3 (LC3) expression, and promoting macrophage autophagy at inflammatory sites. Dictamnine also could reduce the release of interleukin-1β (IL-1β), tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), monocyte chemotactic protein-1 (MCP-1), and interleukin-8 (IL-8), and down-regulate the mRNA expression of these genes in LPS-IFN-γ triggered M1 polarized macrophages. Dictamnine ameliorates AD like skin lesions by inhibiting M1 macrophage polarization and promoting autophagy. Hence, dictamnine is expected to be a potential therapeutic candidate for AD.
Collapse
Affiliation(s)
- Yihan Huang
- School of Pharmacy, Xi'an Jiaotong University
| | | | | | - Yujuan Yuan
- School of Pharmacy, Xi'an Jiaotong University
| | - Ling Gong
- School of Pharmacy, Xi'an Jiaotong University
| | - Rui Liu
- School of Pharmacy, Xi'an Jiaotong University
| | - Jingang An
- Department of Dermatology, the Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University
| |
Collapse
|
144
|
Li J, Wang Y, Tang M, Zhang C, Fei Y, Li M, Li M, Gui S, Guo J. New insights into nanotherapeutics for periodontitis: a triple concerto of antimicrobial activity, immunomodulation and periodontium regeneration. J Nanobiotechnology 2024; 22:19. [PMID: 38178140 PMCID: PMC10768271 DOI: 10.1186/s12951-023-02261-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Accepted: 12/12/2023] [Indexed: 01/06/2024] Open
Abstract
Periodontitis is a chronic inflammatory disease caused by the local microbiome and the host immune response, resulting in periodontal structure damage and even tooth loss. Scaling and root planning combined with antibiotics are the conventional means of nonsurgical treatment of periodontitis, but they are insufficient to fully heal periodontitis due to intractable bacterial attachment and drug resistance. Novel and effective therapeutic options in clinical drug therapy remain scarce. Nanotherapeutics achieve stable cell targeting, oral retention and smart release by great flexibility in changing the chemical composition or physical characteristics of nanoparticles. Meanwhile, the protectiveness and high surface area to volume ratio of nanoparticles enable high drug loading, ensuring a remarkable therapeutic efficacy. Currently, the combination of advanced nanoparticles and novel therapeutic strategies is the most active research area in periodontitis treatment. In this review, we first introduce the pathogenesis of periodontitis, and then summarize the state-of-the-art nanotherapeutic strategies based on the triple concerto of antibacterial activity, immunomodulation and periodontium regeneration, particularly focusing on the therapeutic mechanism and ingenious design of nanomedicines. Finally, the challenges and prospects of nano therapy for periodontitis are discussed from the perspective of current treatment problems and future development trends.
Collapse
Affiliation(s)
- Jiaxin Li
- Department of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China
| | - Yuxiao Wang
- Department of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China
| | - Maomao Tang
- Department of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China
| | - Chengdong Zhang
- Department of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China
| | - Yachen Fei
- Department of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China
| | - Meng Li
- Department of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China
| | - Mengjie Li
- Department of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China.
| | - Shuangying Gui
- Department of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China.
- Institute of Pharmaceutics, Anhui Academy of Chinese Medicine, Hefei, 230012, Anhui, China.
- Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Hefei, 230012, Anhui, China.
- Engineering Technology Research Center of Modernized Pharmaceutics, Anhui Education Department, Hefei, 230012, Anhui, China.
| | - Jian Guo
- Department of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China.
- Institute of Pharmaceutics, Anhui Academy of Chinese Medicine, Hefei, 230012, Anhui, China.
- Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Hefei, 230012, Anhui, China.
- Engineering Technology Research Center of Modernized Pharmaceutics, Anhui Education Department, Hefei, 230012, Anhui, China.
| |
Collapse
|
145
|
Hao L, Zhong X, Yu R, Chen J, Li W, Chen Y, Lu W, Wu J, Wang P. Integrating Network Pharmacology and Experimental Validation to Decipher the Anti-Inflammatory Effects of Magnolol on LPS-induced RAW264.7 Cells. Comb Chem High Throughput Screen 2024; 27:462-478. [PMID: 37818577 DOI: 10.2174/0113862073255964230927105959] [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: 04/14/2023] [Revised: 07/11/2023] [Accepted: 08/11/2023] [Indexed: 10/12/2023]
Abstract
INTRODUCTION Magnolol is beneficial against inflammation-mediated damage. However, the underlying mechanisms by which magnolol exerts anti-inflammatory effects on macrophages remain unclear. OBJECTIVE In this study, network pharmacology and experimental validation were used to assess the effect of magnolol on inflammation caused by lipopolysaccharide (LPS) in RAW264.7 cells. MATERIALS AND METHODS Genes related to magnolol were identified in the PubChem and Swiss Target Prediction databases, and gene information about macrophage polarization was retrieved from the GeneCards, OMIM, and PharmGKB databases. Analysis of protein-protein interactions was performed with STRING, and Cytoscape was used to construct a component-target-disease network. GO and KEGG enrichment analyses were performed to ascertain significant molecular biological processes and signaling pathways. LPS was used to construct the inflammatory cell model. ELISA and qRT.PCR were used to examine the expression levels of inflammationassociated factors, immunofluorescence was used to examine macrophage markers (CD86 and CD206), and western blotting was used to examine protein expression levels. RESULTS The hub target genes of magnolol that act on macrophage polarization were MDM2, MMP9, IL-6, TNF, EGFR, AKT1, and ERBB2. The experimental validation results showed that magnolol treatment decreased the levels of proinflammatory factors (TNF-α, IL-1β, and IL-6). Moreover, the levels of anti-inflammatory factors (IL-10 and IL-4) were increased. In addition, magnolol upregulated the expression of M2 markers (Agr-1, Fizzl, and CD206) and downregulated M1 markers (CD86). The cell experiment results supported the network pharmacological results and demonstrated that magnolol alleviated inflammation by modulating the PI3k-Akt and P62/keap1/Nrf2 signaling pathways. CONCLUSION According to network pharmacology and experimental validation, magnolol attenuated inflammation in LPS-induced RAW264.7 cells mainly by inhibiting M1 polarization and enhancing M2 polarization by activating the PI3K/Akt and P62/keap1/Nrf2 signaling pathways.
Collapse
Affiliation(s)
- Lei Hao
- Department of Surgery Two, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaoying Zhong
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Runjia Yu
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jiahui Chen
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wei Li
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yuzhong Chen
- Department of Surgery Two, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Weiqi Lu
- Department of Surgery Two, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jianyu Wu
- Department of Surgery Two, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Peizong Wang
- State Key Laboratory of Oncology in South China, Department of Anesthesiology, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, 510060, P.R. China
| |
Collapse
|
146
|
Bui I, Bonavida B. Polarization of M2 Tumor-Associated Macrophages (TAMs) in Cancer Immunotherapy. Crit Rev Oncog 2024; 29:75-95. [PMID: 38989739 DOI: 10.1615/critrevoncog.2024053830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/12/2024]
Abstract
We have witnessed in the last decade new milestones in the treatment of various resistant cancers with new immunotherapeutic modalities. These advances have resulted in significant objective durable clinical responses in a subset of cancer patients. These findings strongly suggested that immunotherapy should be considered for the treatment of all subsets of cancer patients. Accordingly, the mechanisms underlying resistance to immunotherapy must be explored and develop new means to target these resistant factors. One of the pivotal resistance mechanisms in the tumor microenvironment (TME) is the high infiltration of tumor-associated macrophages (TAMs) that are highly immunosuppressive and responsible, in large part, of cancer immune evasion. Thus, various approaches have been investigated to target the TAMs to restore the anti-tumor immune response. One approach is to polarize the M2 TAMS to the M1 phenotype that participates in the activation of the anti-tumor response. In this review, we discuss the various and differential properties of the M1 and M2 phenotypes, the molecular signaling pathways that participate in the polarization, and various approaches used to target the polarization of the M2 TAMs into the M1 anti-tumor phenotype. These approaches include inhibitors of histone deacetylases, PI3K inhibitors, STAT3 inhibitors, TLR agonists, and metabolic reprogramming. Clearly, due to the distinct features of various cancers and their heterogeneities, a single approach outlined above might only be effective against some cancers and not others. In addition, targeting by itself may not be efficacious unless used in combination with other therapeutic modalities.
Collapse
Affiliation(s)
- Indy Bui
- University of California Los Angeles
| | - Benjamin Bonavida
- Department of Microbiology, Immunology, & Molecular Genetics, David Geffen School of Medicine at UCLA, Johnson Comprehensive Cancer Center, University of California at Los Angeles, Los Angeles, CA 90025-1747, USA
| |
Collapse
|
147
|
Ma H, Lou K, Shu Q, Song X, Xu H. Aldehyde dehydrogenase 2 deficiency reinforces formaldehyde-potentiated pro-inflammatory responses and glycolysis in macrophages. J Biochem Mol Toxicol 2024; 38:e23518. [PMID: 37638564 DOI: 10.1002/jbt.23518] [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: 11/03/2022] [Revised: 03/05/2023] [Accepted: 08/17/2023] [Indexed: 08/29/2023]
Abstract
Aldehyde dehydrogenase 2 (ALDH2) deficiency caused by genetic variant is present in more than 560 million people of East Asian descent, which can be identified by apparent facial flushing from acetaldehyde accumulation after consuming alcohol. Recent findings indicated that ALDH2 also played a critical role in detoxification of formaldehyde (FA). Our previous studies showed that FA could enhance macrophagic inflammatory responses through the induction of HIF-1α-dependent glycolysis. In the present study, pro-inflammatory responses and glycolysis promoted by 0.5 mg/m3 FA were found in mice with Aldh2 gene knockout, which was confirmed in the primary macrophages isolated from Aldh2 gene knockout mice treated with 50 μM FA. FA at 50 and 100 μM also induced stronger dose-dependent increases of pro-inflammatory responses and glycolysis in RAW264.7 murine macrophages with knock-down of ALDH2, and the enhanced effects induced by 50 μM FA was alleviated by inhibition of HIF-1α in RAW264.7 macrophages with ALDH2 knock-down. Collectively, these results clearly demonstrated that ALDH2 deficiency reinforced pro-inflammatory responses and glycolysis in macrophages potentiated by environmentally relevant concentration of FA, which may increase the susceptibility to inflammation and immunotoxicity induced by environmental FA exposure.
Collapse
Affiliation(s)
- Huijuan Ma
- Shanghai Key Laboratory of New Drug Design, State Key Laboratory of Bioreactor Engineering, School of Pharmacy, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, East China University of Science and Technology, Shanghai, China
| | - Kaiyan Lou
- Shanghai Key Laboratory of New Drug Design, State Key Laboratory of Bioreactor Engineering, School of Pharmacy, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, East China University of Science and Technology, Shanghai, China
| | - Qi Shu
- Shanghai Key Laboratory of New Drug Design, State Key Laboratory of Bioreactor Engineering, School of Pharmacy, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, East China University of Science and Technology, Shanghai, China
| | - Xiaodong Song
- Medical Laboratory Department, Hua Shan Hospital North, Fudan University, Shanghai, China
| | - Huan Xu
- Shanghai Key Laboratory of New Drug Design, State Key Laboratory of Bioreactor Engineering, School of Pharmacy, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, East China University of Science and Technology, Shanghai, China
| |
Collapse
|
148
|
Yasen A, Yang Z, Feng J, Liang R, Dai T, Li K, Cai Y, Wang G. IL-33/ST2 Signaling and its Correlation with Macrophage Heterogeneity and Clinicopathologic Features in Human Intrahepatic Cholangiocarcinoma. Curr Cancer Drug Targets 2024; 24:1144-1156. [PMID: 38299398 DOI: 10.2174/0115680096276605240108112135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 12/15/2023] [Accepted: 12/19/2023] [Indexed: 02/02/2024]
Abstract
BACKGROUND IL-33/ST2 signaling plays crucial roles in the development and progression of various human malignancies. However, its significance in intrahepatic cholangiocarcinoma (ICC) still remains unclear. OBJECTIVE This study aimed to investigate the expression of IL-33/ST2 signaling and its correlations with macrophage heterogeneity and ICC patients' clinicopathologic features. METHODS The expression of different phenotype macrophage markers and IL-33/ST2 signalingrelated markers was detected. The correlation between L-33/ST2 signaling and different phenotype macrophage markers as well as ICC patients' clinicopathologic data was evaluated. RESULTS Massive heterogeneous cancer cells and PAS-positive cells were observed in tumor tissues. CD68-positive cells accumulated in tumor tissues and expression of both M1 phenotype markers and M2 phenotype macrophage markers was higher in tumor samples than para-carcinoma samples. However, M2 phenotype macrophages represented the dominant macrophage population in ICC tissues. Plasma levels of IL-33, ST2, and MIF were evidently enhanced in ICC patients compared to healthy controls. IL-33/ST2 signaling-related markers exhibited a massive increase in tumor samples than para-carcinoma samples. IL-33 and ST2 expression in ICC tissues was positively associated with M1 and M2 phenotype macrophages. Plasma levels of IL-33, ST2, and MIF were correlated with the diameter of tumor lesions, lymph node metastasis, TNM stage, and tumor differentiation degree. Multivariate analysis demonstrated IL-33 expression to exhibit a correlation with the diameter of tumor lesions, lymph node metastasis, and TNM stage. Additionally, there was a relationship observed between ST2, MIF expression, and diameter of tumor lesions plus TNM stage. CONCLUSION IL-33/ST2 signaling exhibited a positive relationship with macrophage heterogeneity in ICC tissues, and upregulated levels of IL-33, ST2, and MIF were associated with aggressive clinicopathologic characteristics. These findings may provide promising diagnostic biomarkers and potential therapeutic strategies for ICC patients targeting IL-33/ST2 signaling.
Collapse
Affiliation(s)
- Aimaiti Yasen
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Army Medical University, No. 183 Xinqiao High Street, Shapingba District, Chongqing, 400037, China
| | - ZhanDong Yang
- Guangzhou National Laboratory, Guangzhou International Bio Island, Guangzhou 510005, Guangdong, Province, China
| | - Jun Feng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, Guangdong Province, China
| | - RunBin Liang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong Province, China
| | - TianXing Dai
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong Province, China
| | - Kai Li
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong Province, China
| | - YuHong Cai
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong Province, China
| | - GuoYing Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong Province, China
| |
Collapse
|
149
|
Wang Y, Ji X, Wang X, Sun M, Li C, Wu D. The injectable hydrogel loading cannabidiol to regulate macrophage polarization in vitro for the treatment of chronic enteritis. J Appl Biomater Funct Mater 2024; 22:22808000241289022. [PMID: 39385453 DOI: 10.1177/22808000241289022] [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] [Indexed: 10/12/2024] Open
Abstract
OBJECTIVE Chronic bowel disease has the characteristics of high recurrence rate, prolonged and non-healing, and the incidence has increased year by year in recent years. Cannabidiol (CBD) has significant anti-inflammatory and antioxidant activities, but it is limited by its characteristics of fat solubility and low bioavailability. This study aims to treat chronic inflammatory bowel disease by preparing a CBD-loaded hydrogel system (GelMA + CBD) that can deliver CBD in situ and improve its bioavailability through slow release. METHOD The study designed and constructed GelMA + CBD, and its surface morphology was observed by scanning electron microscopy, and its pore size, swelling rate and release rate were evaluated to evaluate its bioactivity and biosafety. The expression of various inflammatory factors was detected by ELISA, and the expression of protein and reactive oxygen species were observed by laser confocal microscopy to evaluate their anti-inflammatory and antioxidant properties. RESULTS Our study found that GelMA + CBD with biosafety, could make CBD be slowly released, and effectively inhibit the M1-type polarization of macrophages in vitro, and promote the M2-type polarization. In addition, GelMA + CBD can also reduce the expression of pro-inflammatory factors (such as iNOS) in macrophages, and increase the expression of anti-inflammatory factors (such as Arg-1), clear intracellular reactive oxygen species (ROS), and relieve oxidative stress. CONCLUSION The vitro experiments have confirmed that the CBD-loaded hydrogel system has good biosafety, and can alleviate inflammation by regulating the polarization direction of macrophages, and then inhibiting the secretion of pro-inflammatory factors, laying a strong foundation for the treatment of chronic enteritis.
Collapse
Affiliation(s)
- Ye Wang
- Key Laboratory of Microecology-Immune Regulatory Network and Related Diseases School of Basic Medicine, Jiamusi University, Jiamusi, Heilongjiang Province, China
- Tianjin First Central Hospital, Tianjin, China
| | - Xingming Ji
- Tianjin First Central Hospital, Tianjin, China
- School of Medicine, Nankai University, Tianjin, China
| | - Xinyi Wang
- Tianjin First Central Hospital, Tianjin, China
| | - Mengyu Sun
- Tianjin First Central Hospital, Tianjin, China
| | - Cheng Li
- Tianjin First Central Hospital, Tianjin, China
- School of Medicine, Nankai University, Tianjin, China
| | - Dongmei Wu
- School of Basic Medicine, Jiamusi University, Jiamusi, Heilongjiang, China
| |
Collapse
|
150
|
Hu S, Xu J, Wu M, Yue K, Cui Y, Bai Y, You L, Guo J. Exploration of the mechanism of reinfusion of fresh autologous blood in type 2 diabetes mice to induce macrophage polarization and inhibit erythrocyte damage. J Diabetes Investig 2024; 15:70-77. [PMID: 37846170 PMCID: PMC10759728 DOI: 10.1111/jdi.14089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 06/02/2023] [Accepted: 09/11/2023] [Indexed: 10/18/2023] Open
Abstract
AIMS/INTRODUCTION Type 2 diabetes triggers an inflammatory response that can damage red blood cells. M2 macrophages have inhibitory effects on inflammation, and play an important role in tissue damage repair and fibrosis. Autologous blood transfusion has the potential to inhibit red blood cell damage by mediating macrophage polarization. MATERIALS AND METHODS Swiss mice were used to establish a suitable type 2 diabetes model, and autologous blood transfusion was carried out. The mice were killed, the blood of the mice was collected and CD14+ monocytes were sorted. The expression levels of phenotypic molecules CD16, CD32 and CD206 in CD14+ monocytes were analyzed by flow cytometry. The proportion of M1 and M2 macrophages were analyzed by flow cytometry. The Q value, P50 , 2,3-diphosphoglycerate and Na+ -K+ -ATPase of red blood cells were detected. The red blood cell osmotic fragility test analyzed the red blood cell osmotic fragility. Western blot analysis was used to analyze the expression changes of erythrocyte surface membrane proteins or transporters erythrocyte membrane protein band 4.1, sphingosine-1-phosphate, glycolipid transfer protein and signal peptide peptidase-like 2A. RESULTS Autologous blood transfusion induced a significant increase in the number of macrophages. The state and capacity of blood cells improved with autologous blood transfusion. Reinfusion of fresh autologous blood in type 2 diabetes mice made erythrocytes shrink. The expression of erythrocyte-related proteins proved that the erythrocyte injury in the reinfusion of fresh autologous blood + type 2 diabetes group was significantly reduced. CONCLUSION The reinfusion of fresh autologous blood into the body of patients with type 2 diabetes can induce macrophage polarization to M2, thereby inhibiting red blood cell damage.
Collapse
Affiliation(s)
- She‐jun Hu
- Department of Anesthesiology, Shanghai Gongli HospitalNaval Military Medical UniversityShanghaiChina
| | - Jia‐ming Xu
- Department of Anesthesiology, Shanghai Gongli HospitalNaval Military Medical UniversityShanghaiChina
| | - Man‐di Wu
- Postgraduate training base at Shanghai Gongli HospitalNingxia Medical UniversityYinchuanChina
| | - Ke Yue
- School of MedicineShanghai UniversityShanghaiChina
| | - Ying‐hui Cui
- Department of Anesthesiology, Shanghai Gongli HospitalNaval Military Medical UniversityShanghaiChina
| | - Yu Bai
- Department of Anesthesiology, Shanghai Gongli HospitalNaval Military Medical UniversityShanghaiChina
| | - Lai‐wei You
- Department of Anesthesiology, Shanghai Gongli HospitalNaval Military Medical UniversityShanghaiChina
| | - Jian‐Rong Guo
- Department of Anesthesiology, Shanghai Gongli HospitalNaval Military Medical UniversityShanghaiChina
| |
Collapse
|