1
|
Lou J, Wu X, Ji W, Yu J, Xu Y, Xiao W, Lu W, Xin K, Chen T, Tang Q, Liang G, Gao Y, Wu D. N-Terminal random curl-tandam α-helical peptide 7W: A potent antibacterial and anti-inflammatory dual-effect agent through tryptophan substitution. Eur J Med Chem 2025; 292:117686. [PMID: 40319576 DOI: 10.1016/j.ejmech.2025.117686] [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: 01/10/2025] [Revised: 04/17/2025] [Accepted: 04/24/2025] [Indexed: 05/07/2025]
Abstract
This study investigates the impact of tryptophan substitution on the properties of the Medisin family peptide MS-PT. By substituting hydrophobic amino acids in MS-PT1 with tryptophan, a series of derivative peptides were synthesized. Among them, the 7W peptide stood out with its unique N-terminal random curl and α-helix structure. In vitro, 7W effectively inhibited the secretion of pro-inflammatory cytokines like IL-6 and TNF-α in LPS-induced Membrane-Proximal Macrophages (MPMs) by blocking the MAPK/NF-κB signaling pathway. It also exhibited stronger antimicrobial activity against Gram-positive bacteria compared to the parent peptide MS-PT1, with good safety as indicated by a low hemolysis rate. In vivo, in the CLP-induced sepsis mouse model, 7W alleviated lung and liver injury, suppressed the expression of inflammatory factors in serum and tissues, and had a relatively long plasma half-life of 46.8 h. Mechanistically, 7W interacted preferentially with bacterial mimic membranes and LPS, and its anti-inflammatory effect might be mediated by binding to TLR4. These findings not only clarify the role of tryptophan substitution in modulating peptide properties but also offer a new strategy for the development of multifunctional antimicrobial peptides, suggesting that 7W has great potential as a therapeutic agent for sepsis and other inflammatory diseases.
Collapse
Affiliation(s)
- Jietao Lou
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Xinyi Wu
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China; Department of Pharmacy, The First People's Hospital of Jiande, Hangzhou, 311600, China
| | - Wenwen Ji
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Jiaye Yu
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Yanyan Xu
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Wanyang Xiao
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China; College of Life and Environmental Science, Wenzhou University, Wenzhou, 325035, China
| | - Weijie Lu
- College of Life and Environmental Science, Wenzhou University, Wenzhou, 325035, China
| | - Kaiyun Xin
- College of Life and Environmental Science, Wenzhou University, Wenzhou, 325035, China
| | - Tianbao Chen
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7BL, UK
| | - Qidong Tang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China; Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001, China
| | - Guang Liang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China; School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, 310053, China
| | - Yitian Gao
- College of Life and Environmental Science, Wenzhou University, Wenzhou, 325035, China.
| | - Di Wu
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China.
| |
Collapse
|
2
|
Lei X, Wang F, Zhang X, Huang J, Huang Y. The potential mechanisms by which Xiaoyao Powder may exert therapeutic effects on thyroid cancer were examined at various levels. Comput Biol Chem 2025; 117:108412. [PMID: 40056710 DOI: 10.1016/j.compbiolchem.2025.108412] [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: 12/06/2024] [Revised: 02/13/2025] [Accepted: 02/24/2025] [Indexed: 03/10/2025]
Abstract
BACKGROUND Thyroid cancer (TC) is the most prevalent endocrine malignancy, with a rising incidence necessitating safer treatment strategies to reduce overtreatment and its side effects. Xiaoyao Powder (XYP), a widely used herbal formula, shows promise in treating TC. This study aims to investigate the mechanisms by which XYP may affect TC. METHODS The components of XYP were identified through database retrieval, and targets related to TC were collected to construct a target network for key screening. GEO dataset samples analyzed immune cells and identified significantly differentially expressed core genes (SDECGs). Based on SDECG expression and clustering, samples were classified for comparison. WGCNA was employed to identify gene modules linked to clinical characteristics. ML models screened characteristic genes and constructed a nomogram validated using another GEO dataset. MR methods explored causal relationships between genes and TC. RESULTS The top ten active components of XYP were identified, along with 27 SDECGs that exhibited significant differences in immune cell infiltration between TC patients and normal controls. The nomogram effectively predicted TC risk, validated through ROC curves. Key characteristic genes included SMIM1, PPP1R16A, KIAA1462, DNAJC22, and EFNA5. CONCLUSION XYP may treat TC by regulating SMIM1, PPP1R16A, KIAA1462, DNAJC22, EFNA5, and associated immune pathways; this provides theoretical support for its potential mechanisms.
Collapse
Affiliation(s)
- Xiaoli Lei
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Feifei Wang
- Department of Quality Control, Liaocheng Traditional Chinese Medicine Hospital, Liaocheng, China
| | - Xinying Zhang
- Department of Endocrinology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jiaxi Huang
- Department of Pharmacy, Huoqiu County First People's Hospital, Liuan, China
| | - Yanqin Huang
- Department of Endocrinology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China.
| |
Collapse
|
3
|
Yan K, He Q, Tang J, Peng W, Dou B, Chen H, Bei W. Actinobacillus pleuropneumoniae infection activates IL-1β expression in porcine alveolar macrophages via β-amyloid production. Microb Pathog 2025; 204:107559. [PMID: 40220800 DOI: 10.1016/j.micpath.2025.107559] [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: 12/21/2024] [Revised: 03/30/2025] [Accepted: 04/07/2025] [Indexed: 04/14/2025]
Abstract
Actinobacillus pleuropneumoniae (A. pleuropneumoniae), a porcine respiratory tract pathogen, causes porcine pleuropneumonia. Porcine alveolar macrophages (PAMs) play a crucial role during A. pleuropneumoniae infection. Amyloid precursor protein (APP) can be cleaved by β- and γ-secretase to produce β-amyloid (Aβ). APP and Aβ are associated with the inflammatory response. They activate microglia and astrocytes to secrete IL-1β, IL-6, and other cytokines. In this study, we found that during the interaction between A. pleuropneumoniae and PAMs, the two-component system CpxAR upregulates wecA expression, increasing lipopolysaccharide (LPS) production. LPS promotes APP production and cleavage to generate Aβ. The Aβ activates NF-κB, leading to increased IL-1β expression. We hypothesize that A. pleuropneumoniae infection of PAMs regulates APP production and cleavage to control Aβ levels. Different quantities of Aβ induce PAMs to produce varying amounts of cytokines, leading to different pathological processes in porcine pleuropneumonia.
Collapse
Affiliation(s)
- Kang Yan
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China; The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Qiyun He
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China; The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Jia Tang
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China; The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Wei Peng
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China; The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Beibei Dou
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Huanchun Chen
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China; The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, Hubei, China; Hubei Hongshan Laboratory, Wuhan, China
| | - Weicheng Bei
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China; The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, Hubei, China; Hubei Hongshan Laboratory, Wuhan, China.
| |
Collapse
|
4
|
Xie K, Zhang Y, Ou X, Xiao Y, Luo J, Tan S. Taurine ameliorates liver fibrosis by repressing Fpr2-regulated macrophage M1 polarization. Eur J Pharmacol 2025; 997:177614. [PMID: 40216178 DOI: 10.1016/j.ejphar.2025.177614] [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: 10/30/2024] [Revised: 03/21/2025] [Accepted: 04/08/2025] [Indexed: 04/20/2025]
Abstract
Liver fibrosis is a reversible pathophysiological condition characterized by excessive extracellular matrix deposition that can progress to cirrhosis and liver failure if left untreated. Taurine, a sulfur-containing amino acid, protects the liver from damage. However, the effects of taurine on liver fibrogenesis have not been completely elucidated. In this study, we used amino acid metabolomics, gene expression microanalysis, and single-cell RNA sequencing (scRNA-seq) to investigate the roles of taurine, formyl peptide receptor 2 (Fpr2), and proinflammatory macrophages in liver fibrosis in human fibrotic sections and two distinct mouse models of liver fibrosis. Taurine transporter SLC6A6 wild-type and knockout littermate models and critical element inhibitors were also used. We found that taurine levels were significantly reduced in both human and murine fibrotic sections and that exogenous taurine supplementation alleviated fibrosis via SLC6A6. Furthermore, gene expression microarray analysis and scRNA-seq analyses demonstrated that exogenous taurine mitigated liver fibrosis, mainly by regulating Fpr2-related macrophage status. WRW4-mediated inhibition of Fpr2 ameliorated M1 macrophage polarization and alleviated liver fibrosis. Additionally, exogenous taurine suppressed Fpr2-modulated macrophage M1 polarization and the production of associated proinflammatory cytokines by repressing NF-κBp65 phosphorylation; moreover, SLC6A6 deficiency or treatment of liver fibrosis mouse models with an NF-κB inhibitor, BAY, impaired this protective effect of taurine. Therefore, taurine exerts a protective effect against liver fibrosis by repressing Fpr2/NF-κBp65-regulated macrophage M1 polarization, highlighting its potential therapeutic agent.
Collapse
Affiliation(s)
- Kaiduan Xie
- Department of Gastroenterology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, 510630, China
| | - Yiwang Zhang
- Department of Pathology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, 510630, China
| | - Xingtong Ou
- Department of Gastroenterology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, 510630, China
| | - Yuelin Xiao
- Department of Gastroenterology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, 510630, China
| | - Jiajie Luo
- Department of Gastroenterology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, 510630, China
| | - Siwei Tan
- Department of Gastroenterology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, 510630, China.
| |
Collapse
|
5
|
Ghamangiz S, Jafari A, Maleki-Kakelar H, Azimi H, Mazloomi E. Reprogram to heal: Macrophage phenotypes as living therapeutics. Life Sci 2025; 371:123601. [PMID: 40189197 DOI: 10.1016/j.lfs.2025.123601] [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: 01/15/2025] [Revised: 03/15/2025] [Accepted: 04/01/2025] [Indexed: 04/26/2025]
Abstract
Macrophages represent a crucial cell type within the immune system, exhibiting significant adaptability that allows for the transformation into various phenotypes in response to their surrounding environment. This review investigates the characteristics of various macrophage phenotypes and their functional roles in disease pathogenesis and resolution. The M1 phenotype, recognized for its inflammatory attributes, plays a pivotal role in combating infections and tumors; however, it may also contribute to tissue injury, persistent inflammation, and the pathogenesis of autoimmune and inflammatory diseases. Conversely, the M2 phenotype is associated with anti-inflammatory activities and tissue repair processes. But this is not the end of the story and researches illustrated novel phenotypes that may provide new approaches and therapeutic opportunities. Recent progress in characterizing distinct macrophage phenotypes has enabled the development of innovative therapeutic strategies for chronic inflammatory conditions, autoimmune disorders, and cancers. This review underscores the critical role of macrophage polarization, illustrating how various stimuli can influence macrophage fate and modify their responses. Additionally, it explores the implications of macrophage plasticity on disease progression and treatment efficacy. A comprehensive understanding of these dynamics is essential for the advancement of targeted immunotherapies, which possess the potential to transform treatment strategies for a variety of medical conditions.
Collapse
Affiliation(s)
- Sheyda Ghamangiz
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Abbas Jafari
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Hadi Maleki-Kakelar
- Solid Tumor Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Hadi Azimi
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Ebrahim Mazloomi
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran.
| |
Collapse
|
6
|
Zhang S, Wang Y, Xiong X, Xing J, Jing K. Mechanistic insights into Hippo-YAP pathway activation for enhanced DFU healing. Am J Physiol Cell Physiol 2025; 328:C1921-C1940. [PMID: 40261295 DOI: 10.1152/ajpcell.01067.2024] [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: 01/10/2025] [Revised: 01/16/2025] [Accepted: 04/10/2025] [Indexed: 04/24/2025]
Abstract
With the increasing prevalence of diabetes, diabetic foot ulcers (DFUs) have become a global health challenge, significantly impacting patients' quality of life and placing a substantial burden on healthcare systems. Among various immune cell subsets, M2-polarized macrophages play a pivotal role in tissue repair and inflammation resolution. This study uses single-cell RNA sequencing (scRNA-seq) and bulk RNA sequencing to comprehensively investigate the role of the TFAP2A-LIFR-Hippo-YAP signaling axis in regulating macrophage M2 polarization and its critical function in DFU wound healing. Through scRNA-seq analysis, we identified nine major immune cell subsets in DFU samples, with macrophages emerging as key regulatory cells. In vitro experiments further confirmed that TFAP2A promotes macrophage M2 polarization (evidenced by increased expression of the M2 marker ARG1) and ameliorates endothelial dysfunction by enhancing tube formation, improving migration capacity, and upregulating relevant proteins such as VCAM-1. Moreover, TFAP2A serves as a central regulatory gene for macrophage function in DFU by upregulating LIFR expression and activating the Hippo-YAP signaling pathway, thereby inducing M2 polarization and mitigating endothelial dysfunction. Mouse model experiments further demonstrated that the TFAP2A-LIFR-Hippo-YAP signaling axis accelerates DFU wound healing through the induction of macrophage M2 polarization. This study unveils a novel immunoregulatory role of TFAP2A in DFU and provides a promising therapeutic target for the treatment of chronic diabetic wounds.NEW & NOTEWORTHY This study provides unprecedented insights into diabetic foot ulcer healing by demonstrating the novel immunoregulatory role of the TFAP2A-LIFR-Hippo-YAP signaling axis. Leveraging single-cell and bulk transcriptomics, we identify TFAP2A as a crucial regulator of macrophage M2 polarization, essential for wound healing and angiogenesis. These findings offer valuable mechanistic understanding and present TFAP2A as a promising therapeutic target for improving outcomes in chronic diabetic wounds.
Collapse
Affiliation(s)
- Shaochun Zhang
- Department of Orthopedics, The Central Hospital of Ezhou, Ezhou, People's Republic of China
| | - Ye Wang
- Department of Orthopedics, The Central Hospital of Ezhou, Ezhou, People's Republic of China
| | - Xuesong Xiong
- Department of Endocrinology, The Central Hospital of Ezhou, Ezhou, People's Republic of China
| | - Jili Xing
- Department of Gastroenterology, The Central Hospital of Ezhou, Ezhou, People's Republic of China
| | - Ke Jing
- Department of Endocrinology, The Central Hospital of Ezhou, Ezhou, People's Republic of China
| |
Collapse
|
7
|
Bao L, Wei L, Li X, Jiang X, Duan R, Zhou L, Liu L, Tan W, Huang F, Yang Z. Shuteria involucrata alleviates OVA-induced asthma through the TLR4/NF-κB pathway. Fitoterapia 2025; 183:106520. [PMID: 40180133 DOI: 10.1016/j.fitote.2025.106520] [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: 01/21/2025] [Revised: 03/13/2025] [Accepted: 03/30/2025] [Indexed: 04/05/2025]
Abstract
The roots of Shuteria involucrata (Wall.) Wight & Arn., known as "Tong-qian-ma-huang" in Dai folk medicine in China, are renowned for their efficacy in treating asthma. Despite the use of S. involucrata has been reported as ameliorating respiratory diseases, its mechanism of action remains unclear. This work described the effect of ethyl acetate extract of S. involucrate (SSE) on allergic asthma and its potential mechanism of action was assessed. Ovalbumin was used in vivo to establish a BALB/c asthma mouse model. Lipopolysaccharide was used in vitro to stimulate the human macrophage cell line THP-1 to establish an inflammatory model. In vivo and in vitro experiments showed that SSE effectively ameliorated pathological symptoms and attenuated airway inflammation and cellular inflammation in allergic asthma mice, with a mechanism of action potentially associated to the inhibition of the TLR4/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Lue Bao
- Key Laboratory of Yunnan Provincial Department of Education on Substance Benchmark Research of Ethnic Medicines, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Lisha Wei
- Key Laboratory of Yunnan Provincial Department of Education on Substance Benchmark Research of Ethnic Medicines, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Xiaohong Li
- Key Laboratory of Yunnan Provincial Department of Education on Substance Benchmark Research of Ethnic Medicines, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Xiaoyun Jiang
- Key Laboratory of Yunnan Provincial Department of Education on Substance Benchmark Research of Ethnic Medicines, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Rong Duan
- Key Laboratory of Yunnan Provincial Department of Education on Substance Benchmark Research of Ethnic Medicines, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Lingrui Zhou
- Key Laboratory of Yunnan Provincial Department of Education on Substance Benchmark Research of Ethnic Medicines, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Lu Liu
- Key Laboratory of Yunnan Provincial Department of Education on Substance Benchmark Research of Ethnic Medicines, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Wenhong Tan
- Key Laboratory of Yunnan Provincial Department of Education on Substance Benchmark Research of Ethnic Medicines, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Feng Huang
- School of Chinese Materia Medica and Yunnan Key Laboratory of Southern Medicine Utilization, Yunnan University of Chinese Medicine, Kunming 650500, China.
| | - Zhuya Yang
- Key Laboratory of Yunnan Provincial Department of Education on Substance Benchmark Research of Ethnic Medicines, Yunnan University of Chinese Medicine, Kunming 650500, China.
| |
Collapse
|
8
|
Sun L, Liu Y, Sun Q, Wang G, Du B, Liu B, Gao T, Zhao P, Yang Y, Rong R. Polysaccharides from traditional Chinese medicine and their nano-formulated delivery systems for cancer immunotherapy. Carbohydr Polym 2025; 357:123416. [PMID: 40158963 DOI: 10.1016/j.carbpol.2025.123416] [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: 11/25/2024] [Revised: 02/11/2025] [Accepted: 02/17/2025] [Indexed: 04/02/2025]
Abstract
Cancer immunotherapy has evolved into a new generation strategy in the field of anti-tumor treatment. Polysaccharides derived from Traditional Chinese Medicine (TCM) are gaining recognition as powerful immunomodulators in cancer therapy, noted for their multi-target and multi-pathway actions. Owing to their beneficial properties such as water solubility, biocompatibility, and chemical structure modifiability, TCM polysaccharides can also serve as carriers for hydrophobic drugs in the development of innovative drug delivery systems, enhancing synergistic antitumor effects. In this article, we summarize the diverse mechanisms of immunoregulation by TCM polysaccharides in tumor therapy. The applications of these polysaccharides as both active ingredients and drug carriers within nanodelivery systems for cancer immunotherapy are also introduced. Additionally, extensive research on TCM polysaccharides in clinical settings has been collected. Furthermore, discussions are presented on the development prospects and challenges faced by these polysaccharides in the field of tumor immunotherapy. Our goal is to improve researchers' comprehension of TCM polysaccharides in cancer immunotherapy, providing promising strategies to optimize cancer treatment and benefit diverse patient populations.
Collapse
Affiliation(s)
- Linlin Sun
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Yuting Liu
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Qihui Sun
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Guimei Wang
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Baoxiang Du
- Qingdao Academy of Chinese Medical Sciences, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Bodong Liu
- School of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Tian Gao
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Pan Zhao
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Yong Yang
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China; Collaborative Innovation Center for Antiviral Traditional Chinese Medicine in Shandong Province, Jinan 250355, PR China; Shandong Antiviral Engineering Research Center of Traditional Chinese Medicine, Jinan 250355, PR China.
| | - Rong Rong
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China.
| |
Collapse
|
9
|
Maeda T, Sobajima S, Matsumoto T, Tsubosaka M, Matsushita T, Iwaguro H, Kuroda R. Comparison of short-term clinical outcomes of intra-articular injection of micro-fragmented adipose tissue and stromal vascular fraction cells for knee osteoarthritis treatment: A retrospective single-center cohort study. Regen Ther 2025; 29:91-99. [PMID: 40129683 PMCID: PMC11932757 DOI: 10.1016/j.reth.2025.02.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 02/15/2025] [Accepted: 02/27/2025] [Indexed: 03/26/2025] Open
Abstract
Background Stromal vascular fraction (SVF) cells and micro-fragmented adipose tissue (MFAT) have potential for treating knee osteoarthritis (OA), but their efficacy has not been compared. This study aimed to compare the clinical outcomes of SVF and MFAT for knee OA. We hypothesized that SVF provides stronger short-term effects, while MFAT offers more sustained benefits. Methods A retrospective single-center cohort study was conducted on patients with knee OA, with 36 SVF and 36 MFAT cases selected through propensity score matching between September 2017 and February 2022. Patients with KL grades I-IV varus knee OA, significant pain (VAS ≥40), and functional impairment despite conservative treatments were included. Those with knee trauma, severe bony defects, infections, genu valgus, osteonecrosis, rheumatoid arthritis, or severe deformities were excluded. Clinical outcomes were assessed using the visual analog scale, KOOS, knee range of motion, extension/flexion strength, and MRI T2 mapping. Results SVF and MFAT groups demonstrated significant improvements in VAS (p < 0.01 for both groups). Both groups showed notable improvements in extension angle, extension/flexion muscle strength, and KOOS, with no significant differences between them. However, the MFAT group demonstrated significantly greater improvement in flexion angle compared to the SVF group (p = 0.03). No serious adverse events were reported. T2 mapping showed significant improvements in cartilage quality in both groups, with the MFAT group demonstrating superior improvements in specific lateral regions. Responder rate in SVF group initially improved but declined over time; however, the MFAT group showed sustained improvement from six months onward. Conclusion T2 mapping revealed that MFAT had better cartilage preservation than that of SVF cells in less-loaded areas, with a potentially longer-lasting therapeutic effect. These findings offer important insights for clinicians to tailor treatment strategies based on patient needs and disease progression.
Collapse
Affiliation(s)
- Takuma Maeda
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
- Department of Orthopaedic Surgery, Sobajima Clinic, Higashiosaka, Japan
| | - Satoshi Sobajima
- Department of Orthopaedic Surgery, Sobajima Clinic, Higashiosaka, Japan
| | - Tomoyuki Matsumoto
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Masanori Tsubosaka
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Takehiko Matsushita
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Hideki Iwaguro
- Department of Orthopaedic Surgery, Sobajima Clinic, Higashiosaka, Japan
| | - Ryosuke Kuroda
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| |
Collapse
|
10
|
Li S, Huang Y, Sun Q, Li Y, Xie H, Fu Q. Caspase-1 is critical for mice in the defense against Streptococcus equi subsp. zooepidemicus infection by promoting macrophage phagocytosis. Microb Pathog 2025; 203:107499. [PMID: 40122410 DOI: 10.1016/j.micpath.2025.107499] [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: 07/29/2024] [Revised: 03/11/2025] [Accepted: 03/20/2025] [Indexed: 03/25/2025]
Abstract
Streptococcus equi subsp. zooepidemicus (SEZ) is an important pathogen which is responsible for a wide range of diseases in various species. Macrophages are professional phagocytes that can engulf microorganisms and trigger responses leading to microbial death. Caspase-1 is considered as a proinflammatory factor that mediates antibacterial response to protect hosts from bacteria. Here, we revealed a novel role of Caspase-1 in mice against SEZ. Through both in vitro and in vivo infection assays, we demonstrated that the maturation and secretion of the cytokine IL-1β are critically dependent on Caspase-1 activation. The Caspase-1 deficient mice displayed attenuation of bactericidal activity against SEZ, mainly by decreasing the accumulation of macrophage. In addition to the recruitment of macrophages, deficiency of Caspase-1 also impaired the phagocytosis of SEZ by macrophages. Our study demonstrated that Caspase-1 is critical for mice to defense against SEZ depending on the recruitment and phagocytosis of macrophage.
Collapse
Affiliation(s)
- Shun Li
- School of Animal Science and Technology, Foshan University, Guangdong, 528225, China; Foshan University Veterinary Teaching Hospital, Foshan University, Guangdong, 528225, China
| | - Yunfei Huang
- School of Animal Science and Technology, Foshan University, Guangdong, 528225, China; Foshan University Veterinary Teaching Hospital, Foshan University, Guangdong, 528225, China
| | - Qinqin Sun
- School of Animal Science and Technology, Foshan University, Guangdong, 528225, China; Foshan University Veterinary Teaching Hospital, Foshan University, Guangdong, 528225, China
| | - Yajuan Li
- School of Animal Science and Technology, Foshan University, Guangdong, 528225, China; Foshan University Veterinary Teaching Hospital, Foshan University, Guangdong, 528225, China
| | - Honglin Xie
- School of Animal Science and Technology, Foshan University, Guangdong, 528225, China; Foshan University Veterinary Teaching Hospital, Foshan University, Guangdong, 528225, China.
| | - Qiang Fu
- School of Animal Science and Technology, Foshan University, Guangdong, 528225, China; Foshan University Veterinary Teaching Hospital, Foshan University, Guangdong, 528225, China.
| |
Collapse
|
11
|
Zheng K, Wang S, Deng M, Luo Y, Li W, Zeng L, Wang Y. Mechanisms and Therapeutic Strategies of Macrophage Polarization in Intervertebral Disc Degeneration. JOR Spine 2025; 8:e70065. [PMID: 40371270 PMCID: PMC12077540 DOI: 10.1002/jsp2.70065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 03/03/2025] [Accepted: 03/26/2025] [Indexed: 05/16/2025] Open
Abstract
Background Intervertebral disc degeneration (IVDD) is a leading cause of low back pain (LBP), contributing significantly to global disability and productivity loss. Its pathogenesis involves complex processes, including inflammation, cellular senescence, angiogenesis, fibrosis, neural ingrowth, and sensitization. Emerging evidence highlights macrophages as central immune regulators infiltrating degenerated discs, with macrophage polarization implicated in IVDD progression. However, the mechanisms linking macrophage polarization to IVDD pathology remain poorly elucidated. Methods A comprehensive literature review was conducted by searching major databases (PubMed, Web of Science, and Scopus) for studies published in the last decade (2014-2024). Keywords included "intervertebral disc degeneration," "macrophage polarization," "inflammation," "senescence," and "therapeutic strategies." Relevant articles were selected, analyzed, and synthesized to evaluate the role of macrophage polarization in IVDD. Results Macrophage polarization dynamically influences IVDD through multiple pathways. Pro-inflammatory M1 macrophages exacerbate disc degeneration by amplifying inflammatory cytokines (e.g., TNF-α, IL-1β), promoting cellular senescence, and stimulating abnormal angiogenesis and neural ingrowth. In contrast, anti-inflammatory M2 macrophages may mitigate degeneration by suppressing inflammation and enhancing tissue repair. Therapeutic strategies targeting macrophage polarization include pharmacological agents (e.g., cytokines, small-molecule inhibitors), biologic therapies, gene editing, and physical interventions. Challenges persist, such as incomplete understanding of polarization triggers, lack of targeted delivery systems, and limited translational success in preclinical models. Conclusion Macrophage polarization is a pivotal regulator of IVDD pathology, offering promising therapeutic targets. Future research should focus on elucidating polarization mechanisms, optimizing spatiotemporal control of macrophage phenotypes, and developing personalized therapies. Addressing these challenges may advance innovative strategies to halt or reverse IVDD progression, ultimately improving clinical outcomes for LBP patients.
Collapse
Affiliation(s)
- Kaiyuan Zheng
- Department of Rehabilitation Medicine, Intensive Care MedicineAffiliated Hospital of North Sichuan Medical CollegeNanchongChina
| | - Siyu Wang
- Department of Rehabilitation Medicine, Intensive Care MedicineAffiliated Hospital of North Sichuan Medical CollegeNanchongChina
| | - Meng Deng
- Department of Clinical LaboratoryThe First People's Hospital of GuangyuanGuangyuanChina
| | - Yaomin Luo
- Department of Rehabilitation Medicine, Intensive Care MedicineAffiliated Hospital of North Sichuan Medical CollegeNanchongChina
| | - Wen Li
- Department of Rehabilitation Medicine, Intensive Care MedicineAffiliated Hospital of North Sichuan Medical CollegeNanchongChina
| | - Lianlin Zeng
- Department of Rehabilitation MedicineSuining Central HospitalSuiningChina
| | - Yinxu Wang
- Department of Rehabilitation Medicine, Intensive Care MedicineAffiliated Hospital of North Sichuan Medical CollegeNanchongChina
| |
Collapse
|
12
|
Yu F, Gong Z, Li Y, Naseem DF, Li C, Wen M, Zhao B, Xu Z, Zhang S, Zang R, Wu A, Han Q, Wu S, Li H, Song Y. Association of SIRT6 Expression With Risk of Pneumonitis Induced by Radiotherapy in Cancer Patients. Mol Carcinog 2025; 64:1104-1118. [PMID: 40170513 PMCID: PMC12074565 DOI: 10.1002/mc.23900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 02/06/2025] [Accepted: 02/18/2025] [Indexed: 04/03/2025]
Abstract
Thoracic tumours represent a significant proportion of malignant cancers. While radiotherapy (RT) improves prognosis, it can also lead to side effects such as radiation-induced pneumonitis (RP). Since SIRT6 is involved in DNA repair, energy metabolism and inflammation, this study aims to investigate the expression of SIRT6 in lymphocytes as a potential biomarker and therapeutic target for RP. This study included 170 patients diagnosed with thoracic tumours, all of whom underwent thoracic RT. RP was evaluated and classified as severe RP (SRP) and lower as non-severe RP (NSRP). Analyses were performed using SPSS version 26.0 and the R. Among 170 patients in this study, 124 developed NSRP, and 46 experienced SRP. The univariate analysis showed that SIRT6 expression (cOR, 0.33, 95%CI, 0.18-0.97 before RT and 0.31, 0.19-0.98 after RT), clinical factors, dosimetric parameters and haematological/serological parameters were associated with SRP before and after RT. Our multivariable logistic regression showed that SIRT6 expression was significantly associated with risk of SRP before (aOR, 0.32, 95%CI, 0.15-0.96) and after RT (aOR, 0.32, 95%CI, 0.18-0.99) after adjustment with other confounders. Moreover, the receiver operating characteristic curve analysis revealed that the combined multivariable model exhibited superior predictive capability compared to any single predictor (overall AUC, 0.93, 95%CI, 0.90-0.97 before RT and AUC, 0.91, 95%CI, 0.87-0.96 after RT). The expression of SIRT6 alone or in combination with other risk factors was associated with an increased risk of SRP, suggesting a novel approach for the prevention and treatment of radiation pneumonitis in clinical practice.
Collapse
Affiliation(s)
- Fengyuan Yu
- Department of RadiotherapyQingdao UniversityQingdaoChina
- Department of RadiotherapyThe Affiliated Yantai Yuhuangding Hospital of Qingdao UniversityYantaiChina
| | - Zheng Gong
- Department of RadiotherapyQingdao UniversityQingdaoChina
- Department of RadiotherapyThe Affiliated Yantai Yuhuangding Hospital of Qingdao UniversityYantaiChina
| | - Yuan Li
- Department of RadiologyAffiliated Hospital of Nanjing University of Chinese MedicineNanjingPR China
| | - Danial F. Naseem
- Department of Head and Neck SurgeryMD Anderson Cancer CenterHoustonTexasUSA
| | - Chen Li
- Department of RadiotherapyThe Affiliated Yantai Yuhuangding Hospital of Qingdao UniversityYantaiChina
| | - Miaowei Wen
- Department of RadiotherapyThe Affiliated Yantai Yuhuangding Hospital of Qingdao UniversityYantaiChina
| | - Bingying Zhao
- Department of RadiotherapyThe Affiliated Yantai Yuhuangding Hospital of Qingdao UniversityYantaiChina
| | - Zhezhe Xu
- Department of RadiotherapyThe Affiliated Yantai Yuhuangding Hospital of Qingdao UniversityYantaiChina
| | - Shanshan Zhang
- Department of RadiotherapyThe Affiliated Yantai Yuhuangding Hospital of Qingdao UniversityYantaiChina
| | - Rukun Zang
- Department of RadiotherapyThe Affiliated Yantai Yuhuangding Hospital of Qingdao UniversityYantaiChina
| | - Ailu Wu
- Department of RadiotherapyThe Affiliated Yantai Yuhuangding Hospital of Qingdao UniversityYantaiChina
| | - Qingxin Han
- Department of RadiotherapyThe Affiliated Yantai Yuhuangding Hospital of Qingdao UniversityYantaiChina
| | - Shuhui Wu
- Department of OtorhinolaryngologyBaoshan Hospital Affiliated with Shanghai University of Traditional Chinese MedicineShanghaiPR China
| | - Hongwei Li
- Department of RadiotherapyQingdao UniversityQingdaoChina
| | - Yipeng Song
- Department of RadiotherapyQingdao UniversityQingdaoChina
- Department of RadiotherapyThe Affiliated Yantai Yuhuangding Hospital of Qingdao UniversityYantaiChina
| |
Collapse
|
13
|
Lv Y, Zhang L. IRF7 Activates LCN2 Transcription to Enhance LPS-Induced Acute Lung Injury by Inducing Macrophage Ferroptosis and M1 Polarization. Cell Biochem Biophys 2025; 83:2415-2430. [PMID: 39738844 DOI: 10.1007/s12013-024-01651-9] [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: 09/19/2024] [Accepted: 12/17/2024] [Indexed: 01/02/2025]
Abstract
Acute lung injury (ALI), a severe pulmonary disorder that poses a significant threat to life, is closely associated with macrophage ferroptosis and polarization. Lipocalin 2 (LCN2) has been previously reported to be implicated in the pathogenesis of ALI. However, the specific role of LCN2 in macrophage ferroptosis and polarization remains undetermined. Lipopolysaccharide (LPS) was used to establish a mouse model of ALI and also to stimulate mouse RAW264.7 cells. H&E staining was used for histopathologic evaluation, and immunohistochemistry analysis was used to determine the 4-HNE-positive cells. The secretion levels of TNF-α, IL-6, and IL-1β were assessed by ELISA. Gene and protein expression assays were performed using quantitative PCR and immunoblotting. The levels of MDA, GSH, ROS, and lipid ROS were detected to evaluate the alteration in ferroptosis. CD86+ and CD206+ cells were quantified by flow cytometry. The relationship between LCN2 and interferon regulatory factor 7 (IRF7) was confirmed by chromatin immunoprecipitation (ChIP) and luciferase reporter assays. LCN2 expression was upregulated in the lungs of LPS-induced ALI mice and LPS-stimulated RAW264.7 cells. In LPS-induced ALI mice, the depletion of LCN2 alleviated lung injury and ferroptosis, and also inhibited inflammation and macrophage M1 polarization. In LPS-stimulated RAW264.7 cells, the depletion of LCN2 suppressed ferroptosis, inflammation, and M1 polarization. Mechanistically, IRF7 enhanced LCN2 transcription in RAW264.7 cells by binding to its promoter region. More importantly, the silencing of IRF7 inhibited ferroptosis and M1 polarization in LPS-stimulated RAW264.7 cells by downregulating LCN2. Taken together, the IRF7/LCN2 cascade enhances the ferroptosis and M1 polarization of LPS-stimulated macrophages, thereby exacerbating ALI. Anti-IRF7 and anti-LCN2 therapies might potentially be exploited for the prevention and treatment in ALI.
Collapse
Affiliation(s)
- Yali Lv
- Graduate School, Zhejiang Chinese Medical University, Hangzhou, China
- Emergency Intensive Care Unit (ICU), Jinyun County Second People's Hospital, Lishui, China
| | - Lefeng Zhang
- Department of Respiratory and Critical Care Medicine, Lishui Second People's Hospital, Lishui, China.
| |
Collapse
|
14
|
Li C, Liu J, Feng G, Su J, Xu K, Zhang Z. Circ_0008285 regulates macrophage polarization through miR-375/MAPK14 axis in sepsis-induced acute lung injury. J Mol Histol 2025; 56:168. [PMID: 40418306 DOI: 10.1007/s10735-025-10465-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 05/11/2025] [Indexed: 05/27/2025]
Abstract
Acute lung injury (ALI) induced by sepsis is a serious life-threatening disease, one of its characteristics is the polarization of macrophages. Circ_0008285 has been found to be associated with various diseases. In this study, we detected the regulatory role and mechanism of circ_0008285 in sepsis-induced ALI. RAW264.7 cells treated with LPS and C57BL/6 male mice were used to construct in vitro and in vivo models, respectively. Through A series of experiments such as qRT-PCR, Western blot, CCK-8, flow cytometry, dual-luciferase reporter experiment, HE staining and TUNEL staining, the role of circ_0008285 in sepsis-induced ALI was explored. In LPS-induced RAW264.7 cell, circ_0008285 and MAPK14 were over-expressed, but miR-375 was low-expressed compared with control. The levels of IL-1β, IL-6, TNF-α, iNOS and CD86 were reduced, but CD206 and Arg1 expression were enhanced both in vitro and in vivo after knockdown of circ_0008285. In TC-1 cell co-cultured with LPS+sh-circ_0008285 cells, the viability was increased and the apoptosis level was decreased compared with LPS+sh-NC. Circ_0008285 was the sponge of miR-375, and MAPK14 was the downstream target of miR-375. The injury score, W/D ratio, MPO level and apoptosis level in lung tissue were decreased after knockdown of circ_0008285. Moreover, the total protein, neutrophils and macrophages in BALF were increased. Collectively, this study identified that circ_0008285 could sponge miR-375 to influence MAPK14 expression, and then regulate macrophage polarization of sepsis-induced ALI, which provided new insights for the treatment of sepsis-induced ALI.
Collapse
Affiliation(s)
- Chen Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Hebei North University, No. 12 Changqing Road, Qiaoxi District, Zhangjiakou, 075000, Hebei, China
| | - Jianhua Liu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Hebei North University, No. 12 Changqing Road, Qiaoxi District, Zhangjiakou, 075000, Hebei, China
| | - Gaixia Feng
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Hebei North University, No. 12 Changqing Road, Qiaoxi District, Zhangjiakou, 075000, Hebei, China
| | - Jing Su
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Hebei North University, No. 12 Changqing Road, Qiaoxi District, Zhangjiakou, 075000, Hebei, China
| | - Kailun Xu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Hebei North University, No. 12 Changqing Road, Qiaoxi District, Zhangjiakou, 075000, Hebei, China
| | - Zhihua Zhang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Hebei North University, No. 12 Changqing Road, Qiaoxi District, Zhangjiakou, 075000, Hebei, China.
| |
Collapse
|
15
|
Zhu S, Liu J, Xu K, Xu F, Jiang Y, Dai L, Pei T, Zhu Y, Liu D, Zhang X, Xu J, Yang J, Pan Z, Tao J, Hou Z. Comparative transcriptomic analyses of macrophages infected with Toxoplasma gondii strains of different virulence provide molecular insights into the response of macrophage in phagocytosis and polarization to infection. Mol Immunol 2025; 183:259-273. [PMID: 40414092 DOI: 10.1016/j.molimm.2025.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 03/07/2025] [Accepted: 05/05/2025] [Indexed: 05/27/2025]
Abstract
Macrophages are essential for the proliferation and spread of Toxoplasma gondii. Modulating macrophage activation to improve the inflammatory environment is an effective approach for disease treatment. However, the molecular mechanism through which T. gondii alters macrophage function remain unknown. Based on transcriptomic data analysis of various macrophage types infected with T. gondii, current research revealed differences in the regulation of macrophage functions among strains with different virulence: RH was primarily involved in cell cycle regulation, ME49 was associated with cAMP signaling, and CEP mainly participated in ion channel activity. All three T. gondii strains were involved in regulating immune response activation, including leukocyte adhesion and the MAPK signaling pathway. Nineteen shared DEGs associated with macrophage phagocytosis or polarization were identified through the GeneCards database, and PPI analysis confirmed Il6 as the hub gene in the regulatory network. In vivo and in vitro experiments showed that the YZ-1 strain significantly regulated the expressions of eight DEGs (Il6, Rel, Cd83, Myc, Adora2b, Egr2, Gja1 and Nr4a2), and promoted macrophage phagocytic activity and induced M1 polarization, confirming a significant correlation with Il6. This study revealed the dissimilarities and commonalities in macrophage function regulated by T. gondii strains of different virulence, and identified key molecules involved in the regulation of macrophage phagocytosis and polarization during T. gondii infection. This is crucial for identifying potential drug targets against T. gondii and provides a new perspective on the etiopathogenesis and therapeutic approaches for toxoplasmosis.
Collapse
Affiliation(s)
- Shifan Zhu
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou 225009, PR China; International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, PR China; Jiangsu Key Laboratory of Zoonosis, Yangzhou 225009, PR China
| | - Jiantao Liu
- YEBIO Bioengineering Co., Ltd of QINGDAO, Qingdao 266113, PR China
| | - Kangzhi Xu
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou 225009, PR China; International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, PR China; Jiangsu Key Laboratory of Zoonosis, Yangzhou 225009, PR China
| | - Fan Xu
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou 225009, PR China; International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, PR China; Jiangsu Key Laboratory of Zoonosis, Yangzhou 225009, PR China
| | - Yuwei Jiang
- Lingkou Town Animal Epidemic Prevention Station, Danyang 212353, PR China
| | - Linwei Dai
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou 225009, PR China; International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, PR China; Jiangsu Key Laboratory of Zoonosis, Yangzhou 225009, PR China
| | - Tianxu Pei
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou 225009, PR China; International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, PR China; Jiangsu Key Laboratory of Zoonosis, Yangzhou 225009, PR China
| | - Yuyang Zhu
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou 225009, PR China; International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, PR China; Jiangsu Key Laboratory of Zoonosis, Yangzhou 225009, PR China
| | - Dandan Liu
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou 225009, PR China; International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, PR China; Jiangsu Key Laboratory of Zoonosis, Yangzhou 225009, PR China
| | - Xinjun Zhang
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou 225009, PR China; International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, PR China; Jiangsu Key Laboratory of Zoonosis, Yangzhou 225009, PR China
| | - Jinjun Xu
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou 225009, PR China; International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, PR China; Jiangsu Key Laboratory of Zoonosis, Yangzhou 225009, PR China
| | - Jin Yang
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225000, PR China.
| | - Zhiming Pan
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou 225009, PR China; Jiangsu Key Laboratory of Zoonosis, Yangzhou 225009, PR China
| | - Jianping Tao
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou 225009, PR China; International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, PR China; Jiangsu Key Laboratory of Zoonosis, Yangzhou 225009, PR China.
| | - Zhaofeng Hou
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou 225009, PR China; International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, PR China; Jiangsu Key Laboratory of Zoonosis, Yangzhou 225009, PR China.
| |
Collapse
|
16
|
Xia Y, Tan X, Zeng S, Jiang Y. HRD1 regulates tumor-associated macrophage polarization through USP7 and promotes lung cancer development. Int Immunopharmacol 2025; 159:114944. [PMID: 40414069 DOI: 10.1016/j.intimp.2025.114944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 04/22/2025] [Accepted: 05/20/2025] [Indexed: 05/27/2025]
Abstract
BACKGROUND Lung cancer exhibits high mortality and incidence rates, with tumor-associated macrophages (TAMs) serving as critical contributors to cancer progression. This study investigates the unexplored mechanistic role of HRD1-an E3 ubiquitin ligase implicated in cancer - in orchestrating TAM polarization to affect lung cancer pathogenesis. METHODS HRD1 expression in lung cancer using TCGA database and validated its impact via IHC. THP-1 cells and macrophages isolated from murine tumor tissues via magnetic bead sorting were transfected with the oe-HRD1 plasmid, followed by flow cytometry, ELISA, and RT-qPCR assays to investigate HRD1's regulatory effects on macrophage polarization and function. Co-IP was employed to investigate interactions between USP7 and HRD1/PD-L1, while Immunofluorescence elucidated underlying mechanisms. RESULTS HRD1 was highly expressed in lung cancer and promotes tumor growth in tumor-bearing mice and proliferation in THP-1 cells. Strikingly, both in vivo and in vitro overexpression of HRD1 drove macrophage M2 polarization. Mechanistically, USP7 interacted independently with HRD1 and PD-L1, while HRD1 binding to USP7 facilitated PD-L1 ubiquitination. Furthermore, HRD1 overexpression upregulated USP7 expression, thereby enhancing M2 polarization. CONCLUSION HRD1 promotes lung cancer progression by regulating TAM M2 polarization via USP7, offering novel therapeutic targets and diagnostic perspectives for early-stage lung cancer intervention.
Collapse
Affiliation(s)
- Yezhou Xia
- Pulmonary and Critical Care Medicine, the Second Affiliated Hospital, University of South China, Hengyang, Hunan, China; Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Xiaowu Tan
- Pulmonary and Critical Care Medicine, the Second Affiliated Hospital, University of South China, Hengyang, Hunan, China; Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Saili Zeng
- Pulmonary and Critical Care Medicine, the Second Affiliated Hospital, University of South China, Hengyang, Hunan, China; Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Yixia Jiang
- Pulmonary and Critical Care Medicine, the Second Affiliated Hospital, University of South China, Hengyang, Hunan, China; Hengyang Medical School, University of South China, Hengyang, Hunan, China.
| |
Collapse
|
17
|
Zhang K, Zuo D, Wang Z, Ding J, Xu J, Liu Y, Zhong Y, Jia W. Heterologous prime-boost with an mRNA vaccine and an oncolytic virus enhances tumor regression through overcoming intratumoral immune suppression. Cell Rep 2025; 44:115745. [PMID: 40411783 DOI: 10.1016/j.celrep.2025.115745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 02/10/2025] [Accepted: 05/06/2025] [Indexed: 05/26/2025] Open
Abstract
Therapeutic mRNA vaccines are limited in inducing tumor shrinkage in advanced cancers due to their inability to overcome immune-suppressive mechanisms within tumors. In this study, we developed an HPV-immunogen-expressing oncolytic virus (OV) using HSV-1 for HPV-related cancer treatment. A mouse syngeneic tumor model evaluates the effectiveness of intratumoral OV application for E6+E7+ tumors. Comparative analysis of OV and mRNA vaccines reveals distinct mechanisms in tumor treatment. Single-cell RNA sequencing and flow cytometry show that OV enhances cytotoxic T cell infiltration, polarizes neutrophils and macrophages toward anti-tumor phenotypes, and promotes immune activation within the tumor. In contrast, the mRNA vaccine more effectively activates peripheral antigen-specific T cell responses. A heterologous prime-boost strategy using the mRNA vaccine to prime systemic T cells, followed by OV therapy to direct these cells into the tumor, leads to significant tumor regression. This combination optimizes both systemic and intratumoral immune responses for advanced HPV-related cancers.
Collapse
Affiliation(s)
- Kuan Zhang
- Shanghai Virogin Biotech Co., Ltd., Shanghai 201802, China
| | - Dong Zuo
- Shanghai Virogin Biotech Co., Ltd., Shanghai 201802, China
| | - Zhenglong Wang
- Shanghai Virogin Biotech Co., Ltd., Shanghai 201802, China
| | - Jun Ding
- Shanghai Virogin Biotech Co., Ltd., Shanghai 201802, China; Virogin Biotech Co., Ltd, Richmond, BC V6V 3A4, Canada
| | - Jiang Xu
- Shanghai Virogin Biotech Co., Ltd., Shanghai 201802, China
| | - Yin Liu
- Shanghai Virogin Biotech Co., Ltd., Shanghai 201802, China
| | - Yu Zhong
- Shanghai Virogin Biotech Co., Ltd., Shanghai 201802, China
| | - William Jia
- Shanghai Virogin Biotech Co., Ltd., Shanghai 201802, China; Virogin Biotech Co., Ltd, Richmond, BC V6V 3A4, Canada.
| |
Collapse
|
18
|
Qu Y, Gu Y, Zhang X, Wang Y, Xing X. Acupuncture's Immunomodulatory Effects on Macrophages in Allergic Disorders: A Systematic Review. J Asthma Allergy 2025; 18:801-815. [PMID: 40421260 PMCID: PMC12105631 DOI: 10.2147/jaa.s516732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 05/05/2025] [Indexed: 05/28/2025] Open
Abstract
The incidence of allergic diseases has been increasing annually, severely affecting the quality of life of patients. With the growing recognition of traditional medicine, acupuncture, an ancient Chinese therapeutic method, has gradually gained attention for its potential in immune modulation. Studies have shown that macrophages play a crucial role in the development of allergic diseases, and acupuncture may influence allergic reactions by modulating the function of macrophages. This article aims to systematically evaluate the regulatory effects of acupuncture on macrophages in allergic diseases and the corresponding mechanisms. It analyzes existing research findings and explores the clinical application prospects of acupuncture in this context. By understanding how acupuncture affects the activation, secretion, and role of macrophages in immune responses, we hope to provide new insights and directions for the treatment of allergic diseases.
Collapse
Affiliation(s)
- Yang Qu
- The Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, 150001, People’s Republic of China
| | - Yunhe Gu
- The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150006, People’s Republic of China
| | - Xiaoying Zhang
- The Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, 150001, People’s Republic of China
| | - Yanlong Wang
- The second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150086, People’s Republic of China
| | - Xueliang Xing
- The Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, 150001, People’s Republic of China
| |
Collapse
|
19
|
Guo Z, Shen Y, Yu X, Song Y, Zheng J, Zeng Y, Wang Y, Fu Z, Hou Y, Shi D, Han L, Li J, Chen L. Inhibition of IRE1α Alleviates Renal Fibrosis and Downregulates M1 Macrophage Activation via the p38 MAPK Pathway. Immunology 2025. [PMID: 40405453 DOI: 10.1111/imm.13949] [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: 12/18/2024] [Revised: 04/21/2025] [Accepted: 05/10/2025] [Indexed: 05/24/2025] Open
Abstract
The enhanced M1 macrophage activation and proportion significantly promote the progression of renal fibrosis in the unilateral ureteral obstruction (UUO) model, while the underlying mechanisms need to be further studied. Here, we examined whether or not endoplasmic reticulum (ER) stress contributed to M1 macrophage activation and the mechanisms in this process. In the UUO mouse model, the proportion of M1 macrophages could be significantly increased in the early renal fibrosis, with the ER stress activated. The inhibitor of ER stress (4-PBA) significantly suppressed the activation of M1 macrophages and alleviated the renal fibrosis in the UUO mouse model. Furthermore, the renal fibrosis could be relieved after the administration of IRE1α inhibitor (4μ8C), with the downregulation of ER stress and M1 macrophage activation. Mechanistically, ER stress-enhanced activation of M1 macrophages was regulated through the IRE1α/XBP1s-p38 MAPK pathway. IRE1α-deficient macrophages could alleviate the renal fibrosis in the UUO mouse model. Thus, our findings suggest that the ER stress pathway regulates M1 macrophage activation in the UUO model, which provides a novel therapeutic approach for renal fibrosis.
Collapse
Affiliation(s)
- Zichan Guo
- Faculty of Life Sciences, Northwest University, Xi'an, China
- Department of Immunology, Fourth Military Medical University, Xi'an, China
| | - Yuting Shen
- Department of Immunology, Fourth Military Medical University, Xi'an, China
- Department of Clinical Laboratory, Xi'an People's Hospital (Xi'an Fourth Hospital), Affiliated People's Hospital of Northwest University, Xi'an, China
| | - Xiaxia Yu
- Department of Immunology, Fourth Military Medical University, Xi'an, China
| | - Yun Song
- Department of Immunology, Fourth Military Medical University, Xi'an, China
| | - Jiyang Zheng
- Department of Immunology, Fourth Military Medical University, Xi'an, China
- School of Medicine, Northwest University, Xi'an, China
| | - Yuen Zeng
- Department of Immunology, Fourth Military Medical University, Xi'an, China
| | - Yazhen Wang
- Department of Immunology, Fourth Military Medical University, Xi'an, China
| | - Zhaoyue Fu
- Department of Immunology, Fourth Military Medical University, Xi'an, China
| | - Yongli Hou
- Department of Immunology, Fourth Military Medical University, Xi'an, China
| | - Dingwen Shi
- Department of Immunology, Fourth Military Medical University, Xi'an, China
| | - Liangjian Han
- Department of Immunology, Fourth Military Medical University, Xi'an, China
| | - Juan Li
- Department of Immunology, Fourth Military Medical University, Xi'an, China
| | - Lihua Chen
- Department of Immunology, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
20
|
Pan J, Li Z, Zhu M, Guo L, Chen W, Yu L. Vitamin E exerts a mitigating effect on LPS-induced acute lung injury by regulating macrophage polarization through the AMPK/NRF2/NF-κB pathway. Int Immunopharmacol 2025; 159:114893. [PMID: 40403505 DOI: 10.1016/j.intimp.2025.114893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Revised: 04/28/2025] [Accepted: 05/14/2025] [Indexed: 05/24/2025]
Abstract
Acute Lung Injury (ALI) and its severe manifestation, acute respiratory distress syndrome (ARDS), are major threats to human health, characterized by high mortality rates and a lack of effective treatments. Given the significant role of an over-activated inflammatory response and macrophage polarization in the development of ALI, and the unknown effect of vitamin E in this context, our study aimed to explore vitamin E's potential in alleviating ALI. We established an ALI mouse model by intratracheal instillation of LPS and isolated BMDMs for in-vitro experiments. Results indicated that vitamin E treatment significantly reduced LPS-induced lung injury, as shown by decreased lung wet/dry weight ratio, lower levels of pro-inflammatory factors in bronchoalveolar lavage fluid, and improved mouse survival rates. Vitamin E also alleviated oxidative stress by modulating oxidative and reducing products. Mechanistically, it activated the AMPK signal, upregulated NRF2, scavenged reactive oxygen species, inhibited the NF-κB signal pathway, and regulated macrophage polarization towards the anti-inflammatory M2 phenotype while suppressing the pro-inflammatory M1 polarization. In conclusion, vitamin E may serve as a potential adjuvant treatment for ALI through the AMPK/NRF2 signaling axis, although further research on optimal dosage and combination therapies is needed.
Collapse
Affiliation(s)
- Jianwei Pan
- Department of Pediatrics, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Zhongyue Li
- Department of Pediatrics, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Menglu Zhu
- Department of Pediatrics, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Lina Guo
- Department of Pediatrics, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Wangxing Chen
- Department of Pediatrics, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Lin Yu
- Department of Pediatrics, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China; Department of Pediatric Pulmonology, Children's Medical Center, Peking University First Hospital, No.5 Le Yuan Road, Daxing District, 102600 Beijing, China.
| |
Collapse
|
21
|
Wang H, Wang Y, Ling M, Wang S, Luo J, Sun J, Xi Q, Chen T, Zhang Y. Comparison of the Immunomodulatory Roles of Porcine Milk-Derived Extracellular Vesicles from Different Stages of Lactation. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:12247-12260. [PMID: 40327365 DOI: 10.1021/acs.jafc.4c11983] [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: 05/07/2025]
Abstract
Milk-derived extracellular vesicles (MEV) have received increasing attention due to their physiological benefits for newborn growth and development. However, their physiological characteristics and immunomodulatory capacities during lactation remain unclear. In this study, we isolated MEV from porcine milk at day 1, week 1, week 2, and week 3 after parturition and compared their differences by common EV characterization, showing the higher concentration and smaller mean size of MEV from colostrum. Porcine MEV could be internalized by RAW264.7 macrophages and functionally deliver immune-related microRNAs (miRNAs). Furthermore, MEV promoted macrophage polarization toward the M2-like phenotype and exerted anti-inflammatory effects in vitro. Mice models with sepsis also displayed that MEV suppressed the secretion of pro-inflammatory cytokines in serum and drove an anti-inflammatory M2-like phenotype of the spleen, ultimately weakening immune responses. Especially, MEV from colostrum exhibited the most robust effects on immunoregulation. Overall, this study provides valuable information for MEV secretion patterns and their differences in immunomodulatory bioactivities.
Collapse
Affiliation(s)
- Hailong Wang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Yuxuan Wang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Mingwang Ling
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Shumeng Wang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Junyi Luo
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Jiajie Sun
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Qianyun Xi
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Ting Chen
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Yongliang Zhang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| |
Collapse
|
22
|
Yi X, Yue J, Yue S, Li J, Zhang X, Zhao G, Tang J, Chen J, Huang N, Cheng Y. The Evolution and Recurrence of Chronic Subdural Hematoma was Associated with Different Distribution of Macrophage M1/M2 Polarization. Inflammation 2025:10.1007/s10753-025-02318-0. [PMID: 40397352 DOI: 10.1007/s10753-025-02318-0] [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: 01/12/2025] [Revised: 04/19/2025] [Accepted: 05/13/2025] [Indexed: 05/22/2025]
Abstract
The pathogenesis of chronic subdural hematoma (CSDH) has traditionally been associated with inflammation, but the efficacy of anti-inflammatory drug therapy is limited. Recent literatures suggest that immune dysregulation rather than sole inflammation, plays a vital role in the development of CSDH. In this study, the dynamics of macrophage polarization were explored to elucidate changes in immune status during CSDH evolution. Nakaguchi computerized tomography (CT) classification method was employed to categorize CSDH into four types, including homogeneous, laminar, separated and trabecular subtypes. Samples from the hematoma cavity were collected. The percentages of M1 and M2 macrophages and the M1/M2 proportion were determined by flow cytometry. The M1-related inflammatory (IL-1β, IL-6, IL-12, and TNF-α) and M2-related anti-inflammatory factors (IL-4, IL-10, IL-13, and TGF-β) were measured by ELISA. The relationship among CT subtypes, macrophage polarization and recurrence were evaluated by univariate and multivariate logistic regression analyses. A nomogram was established to score significant factors, and the bootstrap method was used for internal validation to calculate the concordance index (C-index) and generating calibration plots. A total of 127 patients with CSDH were included, among which 28 cases (22.04%) experienced recurrence within three months post-surgery. Significant differences were found in the percentages of M1 and M2 macrophages, the M1/M2 ratio, and related cytokines among different subtypes in CT classifications (P < 0.001). As CSDH evolved according to different CT subtypes, M1 macrophages gradually decreased, while M2 macrophages significantly increased, accompanied by a downregulation of the M1/M2 ratio. The similar changes were found in M1-related inflammatory cytokines (IL-1β, IL-6, IL-12, and TNF-α) and M2-related anti-inflammatory cytokines. Additionally, compared to the non-recurrence group, the recurrence group had higher percentages of M1 and M1/M2 ratio, but lower percentages of M2 (P < 0.05). LASSO regression analysis identified the dichotomized Nakaguchi CT classification, degree of brain atrophy, postoperative drainage volume, and hematoma volume were independent risk factors of recurrence (P < 0.05). Based on this, the established nomogram prediction model showed an AUC of 0.898, indicating good predictive efficacy and accuracy. The study demonstrated that the immune status within the hematoma cavity shifts from an inflammatory to an anti-inflammatory state during CSDH progression. Furthermore, it was found that altered immune balance gives rise to CSDH evolution and recurrence following surgery instead of inflammation itself.
Collapse
Affiliation(s)
- Xiaoyao Yi
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, 74 Linjiang Rd, Yuzhong, 400010, Chongqing, China
| | - Jianhe Yue
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, 74 Linjiang Rd, Yuzhong, 400010, Chongqing, China
| | - Shengtao Yue
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, 74 Linjiang Rd, Yuzhong, 400010, Chongqing, China
| | - Jilai Li
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, 74 Linjiang Rd, Yuzhong, 400010, Chongqing, China
| | - Xiang Zhang
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, 74 Linjiang Rd, Yuzhong, 400010, Chongqing, China
| | - Guanjian Zhao
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, 74 Linjiang Rd, Yuzhong, 400010, Chongqing, China
| | - Jun Tang
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, 74 Linjiang Rd, Yuzhong, 400010, Chongqing, China
| | - Jin Chen
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, 74 Linjiang Rd, Yuzhong, 400010, Chongqing, China
| | - Ning Huang
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, 74 Linjiang Rd, Yuzhong, 400010, Chongqing, China.
| | - Yuan Cheng
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, 74 Linjiang Rd, Yuzhong, 400010, Chongqing, China.
| |
Collapse
|
23
|
Fu X, Zhong C, Liu W, Ren F, Liang C. Development of Injectable Aldehyde Hyaluronic Acid Hydrogels Loaded with CRISPRa Reprogrammed Elite Macrophages for the Treatment of Osteoarthritis. ACS APPLIED MATERIALS & INTERFACES 2025. [PMID: 40397763 DOI: 10.1021/acsami.5c04355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2025]
Abstract
Osteoarthritis (OA) is a common joint disorder that causes significant disability. Previous studies suggested that the predominance of M1 macrophages (MΦs) exacerbates inflammation and cartilage degradation in OA, suggesting that shifting the polarization toward M2 MΦs could be a promising therapeutic strategy. We recently developed CRISPRa-engineered macrophages, termed Elite MΦs, that express IL-10 and maintain a stable M2 phenotype. However, achieving effective and sustained delivery of these cells to the OA joint remains a challenge. In this study, we synthesized two injectable aldehyde hyaluronic acid-based hydrogels, CHO/CDH and ACHO/CDH hydrogels, to serve as Elite MΦ delivery platforms. Comprehensive analyses identified the ACHO/CDH hydrogel as superior due to its enhanced suitability for encapsulating and delivering Elite MΦs. When loaded with Elite MΦs, the ACHO/CDH hydrogel was able to not only localize Elite MΦs but also enhance their anti-inflammatory and reparative effects. Furthermore, intra-articular injection of the Elite MΦ-loaded ACHO/CDH hydrogel in an OA mouse model resulted in notable improvements in the joint's cellular environment, alleviating cartilage degradation and synovial inflammation. These results highlight the ability of the ACHO/CDH hydrogel to rebalance the inflammatory imbalance and promote cartilage repair. This approach not only targets the underlying inflammatory processes more directly than traditional therapies but also harnesses the regenerative potential of macrophages, offering a transformative strategy for OA management.
Collapse
Affiliation(s)
- Xuekun Fu
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Chuanxin Zhong
- Department of Materials Science and Engineering, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Wentao Liu
- Department of Materials Science and Engineering, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Fuzeng Ren
- Department of Materials Science and Engineering, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Chao Liang
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
- Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR 999077, China
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 100850, China
| |
Collapse
|
24
|
Wei G, Li X, Huang M, Wang H, Li Z, Qu S. Light-Responsive Oxygen Generation from Chlorella Hydrogels for Facial Nerve Injury Recovery: Crosstalk between M1/M2 Macrophages and Schwann Cells. Adv Healthc Mater 2025:e2501123. [PMID: 40394959 DOI: 10.1002/adhm.202501123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2025] [Revised: 04/20/2025] [Indexed: 05/22/2025]
Abstract
Facial nerve injury (FNI), hindered by hypoxic microenvironments limiting Schwann cell (SCs) repair potential, remains a therapeutic challenge. We developed light-responsive Chlorella hydrogels (C-Gel) to modulate oxygen release and inflammation. In vitro, light-activated C-Gel enhanced RSC96 SC proliferation, migration, and secretion while reducing reactive oxygen species (ROS), hypoxia-inducible factor-1α (HIF-1α), tumor necrosis factor-alpha (TNF-α), and interleukin-6 (IL-6). It also shifted macrophage polarization from pro-inflammatory M1 (inducible nitric oxide synthase (iNOS)+/TNF-α+) to anti-inflammatory M2 (arginase-1 (Arg-1)+/IL-10+), with M2-conditioned mediumboosting SCs production of neurotrophic factors (nerve growth factor, NGF; glial cell line-derived neurotrophic factor, GDNF), adhesion molecules (neural cell adhesion molecule-1, NCAM-1), regeneration-associated proteins (c-JUN), and myelin components (myelin basic protein, MBP; myelin-associated glycoprotein, MAG). In vivo, C-Gel-light therapy improved behavioral recovery in FNI rats, suppressed inflammation (ROS/HIF-1α/TNF-α), and enhanced SC-mediated remyelination (S100 calcium-binding protein, S100; neurofilament 200, NF200). RNA sequencing identified upregulated phosphoinositide 3-kinase-protein kinase (PI3K-Akt) and calcium ion (Ca²+) signaling pathways. This oxygen-regulating, immunomodulatory biomaterial offers a dual-action strategy to advance FNI rehabilitation by synergistically optimizing the regenerative microenvironment.
Collapse
Affiliation(s)
- Guihua Wei
- School of Life Science and Engineering, Southwest Jiao Tong University, Chengdu, Sichuan, 610031, China
| | - Xi Li
- School of Life Science and Engineering, Southwest Jiao Tong University, Chengdu, Sichuan, 610031, China
| | - Mengqi Huang
- School of Life Science and Engineering, Southwest Jiao Tong University, Chengdu, Sichuan, 610031, China
| | - Haoyan Wang
- School of Life Science and Engineering, Southwest Jiao Tong University, Chengdu, Sichuan, 610031, China
| | - Zaiqi Li
- School of Life Science and Engineering, Southwest Jiao Tong University, Chengdu, Sichuan, 610031, China
| | - Shuxin Qu
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiao tong University, Chengdu, Sichuan, 610031, China
| |
Collapse
|
25
|
Ren Y, Zhou M, Li Y, Li Y, Xiang J, Deng F, Luo Z, Liu E, Yu J, Fu Z, Ding F, Liu B. Exosomes derived from M2 macrophages regulate airway inflammation by modulating epithelial cell proliferation and apoptosis. J Inflamm (Lond) 2025; 22:19. [PMID: 40390011 PMCID: PMC12090695 DOI: 10.1186/s12950-025-00444-y] [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: 02/05/2025] [Accepted: 05/12/2025] [Indexed: 05/21/2025] Open
Abstract
BACKGROUND Asthma is a chronic inflammatory disease characterized by airway remodeling and immune dysregulation. This study aimed to explore the mechanisms by which M2 macrophage-derived exosomes (M2Φ-Exos) regulate airway inflammation in asthma by modulating epithelial cell proliferation and apoptosis. METHODS M2Φ-Exos were extracted and characterized by morphology, size, and marker protein expression. In vitro, the effects of M2Φ-Exos on House Dust Mites (HDM)-stimulated mouse lung epithelial cells (MLE-12s) were evaluated using western blotting to analyze Proliferating Cell Nuclear Antigen (PCNA), B-cell lymphoma-2 (Bcl-2), Bcl-2-associated X protein (Bax), and cleaved caspase-3 expression. In vivo, M2Φ-Exos were administered to HDM-induced asthmatic mice to assess their impact on airway inflammation, epithelial remodeling, and proliferation-apoptosis balance using immunohistochemistry, immunofluorescence, and western blotting. Cytokine levels in lung tissue and bronchoalveolar lavage fluid (BALF) were measured by qRT-PCR and ELISA. RESULTS M2Φ-Exos displayed typical cup-shaped morphology, an average diameter of 115.5 nm, and expressed marker proteins CD9, TSG101, and CD63. MLE-12 cells internalized M2Φ-Exos, leading to reduced abnormal proliferation and apoptosis in HDM-stimulated cells. In asthmatic mice, M2Φ-Exos alleviated airway inflammation and epithelial thickening while reducing PCNA, cleaved caspase-3, and Bax levels and increasing Bcl-2 expression. M2Φ-Exos suppressed pro-inflammatory cytokines (IL-4, IL-5, IL-13) and Transforming growth factor (TGF)-β, while enhancing anti-inflammatory cytokine IFN-γ and IL-10. CONCLUSION These findings demonstrate that M2Φ-Exos regulate the imbalance in epithelial proliferation and apoptosis in asthma, reducing inflammation and mitigating tissue remodeling, and provide new insights into potential therapeutic strategies for asthma management.
Collapse
Affiliation(s)
- Yinying Ren
- Department of Respiratory Medicine, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation base of Child development and Critical Disorders, Chongqing Engineering Research Center of Stem Cell Therapy, Children's Hospital of Chongqing Medical University, No. 136, Zhongshan 2nd Road, Yuzhong Dis, Chongqing, 400014, China
| | - Mi Zhou
- Department of Respiratory Medicine, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation base of Child development and Critical Disorders, Chongqing Engineering Research Center of Stem Cell Therapy, Children's Hospital of Chongqing Medical University, No. 136, Zhongshan 2nd Road, Yuzhong Dis, Chongqing, 400014, China
| | - Yuehan Li
- Department of Respiratory Medicine, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation base of Child development and Critical Disorders, Chongqing Engineering Research Center of Stem Cell Therapy, Children's Hospital of Chongqing Medical University, No. 136, Zhongshan 2nd Road, Yuzhong Dis, Chongqing, 400014, China
| | - Yan Li
- Department of Respiratory Medicine, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation base of Child development and Critical Disorders, Chongqing Engineering Research Center of Stem Cell Therapy, Children's Hospital of Chongqing Medical University, No. 136, Zhongshan 2nd Road, Yuzhong Dis, Chongqing, 400014, China
| | - JinYing Xiang
- Department of Respiratory Medicine, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation base of Child development and Critical Disorders, Chongqing Engineering Research Center of Stem Cell Therapy, Children's Hospital of Chongqing Medical University, No. 136, Zhongshan 2nd Road, Yuzhong Dis, Chongqing, 400014, China
| | - Fang Deng
- Department of Respiratory Medicine, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation base of Child development and Critical Disorders, Chongqing Engineering Research Center of Stem Cell Therapy, Children's Hospital of Chongqing Medical University, No. 136, Zhongshan 2nd Road, Yuzhong Dis, Chongqing, 400014, China
| | - Zhengxiu Luo
- Department of Respiratory Medicine, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation base of Child development and Critical Disorders, Chongqing Engineering Research Center of Stem Cell Therapy, Children's Hospital of Chongqing Medical University, No. 136, Zhongshan 2nd Road, Yuzhong Dis, Chongqing, 400014, China
| | - Enmei Liu
- Department of Respiratory Medicine, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation base of Child development and Critical Disorders, Chongqing Engineering Research Center of Stem Cell Therapy, Children's Hospital of Chongqing Medical University, No. 136, Zhongshan 2nd Road, Yuzhong Dis, Chongqing, 400014, China
| | - Jinyue Yu
- Bristol Medical School, University of Bristol, Bristol, UK
- Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Zhou Fu
- Department of Respiratory Medicine, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation base of Child development and Critical Disorders, Chongqing Engineering Research Center of Stem Cell Therapy, Children's Hospital of Chongqing Medical University, No. 136, Zhongshan 2nd Road, Yuzhong Dis, Chongqing, 400014, China
| | - Fengxia Ding
- Department of Respiratory Medicine, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation base of Child development and Critical Disorders, Chongqing Engineering Research Center of Stem Cell Therapy, Children's Hospital of Chongqing Medical University, No. 136, Zhongshan 2nd Road, Yuzhong Dis, Chongqing, 400014, China.
| | - Bo Liu
- Department of Cardiothoracic Surgery, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation base of Child development and Critical Disorders, Chongqing Engineering Research Center of Stem Cell Therapy, Children's Hospital of Chongqing Medical University, No. 136, Zhongshan 2nd Road, Yuzhong Dis, Chongqing, 400014, China.
| |
Collapse
|
26
|
Kirchhoff R, Chromik MA, Schebb NH. Phagocytosis is differentially regulated by LPS in M1- and M2-like macrophages via PGE 2 formation and EP4 signaling. Prostaglandins Other Lipid Mediat 2025; 178:106998. [PMID: 40383415 DOI: 10.1016/j.prostaglandins.2025.106998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 04/30/2025] [Accepted: 05/15/2025] [Indexed: 05/20/2025]
Abstract
Phagocytosis is a key process in human innate immune response. Human macrophages are important phagocytes engulfing and neutralizing pathogens and cell debris. In addition, they modulate the inflammatory process by releasing cytokines and lipid mediators. However, the link between oxylipins and phagocytosis in different macrophage phenotypes remains poorly understood. In order to better understand the link between phagocytosis and the arachidonic acid (ARA) cascade, we established a phagocytosis assay in primary human 'inflammatory' M1- and 'anti-inflammatory' M2-like macrophages from peripheral blood mononuclear cells (PBMC), representing extremes of macrophage phenotypes. The branches of the ARA cascade were investigated by quantitative targeted proteomics and metabolomics. M1-like macrophages show a higher abundance of cyclooxygenase (COX)-2 and its products particularly after LPS stimulus compared to M2-like macrophages. LPS increased phagocytosis in M2-like, but not in M1-like macrophages. We demonstrate that the COX product prostaglandin E2 (PGE2) modulates the differential effects of LPS on phagocytosis: Via the EP4 receptor PGE2 signaling suppresses phagocytosis in primary human macrophages. Thus, blockage of COX, e.g. by non-steroidal anti-inflammatory drugs (NSAID), leads to an increase of phagocytosis also in 'inflammatory' M1-like macrophages. This supports the well-described anti-inflammatory effects of these drugs and underscores the importance of the link between the COX branch of the ARA cascade and the regulation of phagocytosis in human macrophages.
Collapse
Affiliation(s)
- Rebecca Kirchhoff
- Chair of Food Chemistry, School of Mathematics and Natural Sciences, University of Wuppertal, Gaussstr. 20, Wuppertal 42119, Germany
| | - Michel André Chromik
- Chair of Food Chemistry, School of Mathematics and Natural Sciences, University of Wuppertal, Gaussstr. 20, Wuppertal 42119, Germany
| | - Nils Helge Schebb
- Chair of Food Chemistry, School of Mathematics and Natural Sciences, University of Wuppertal, Gaussstr. 20, Wuppertal 42119, Germany.
| |
Collapse
|
27
|
Wang Y, Zhang H, Miao C. Unraveling immunosenescence in sepsis: from cellular mechanisms to therapeutics. Cell Death Dis 2025; 16:393. [PMID: 40379629 DOI: 10.1038/s41419-025-07714-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Revised: 04/26/2025] [Accepted: 05/02/2025] [Indexed: 05/19/2025]
Abstract
Sepsis is a life-threatening multiple organ dysfunction resulting from a dysregulated host response to infection, and patients with sepsis always exhibit a state of immune disorder characterized by both overwhelming inflammation and immunosuppression. The aging of immune system, namely "immunosenescence", has been reported to be correlated with high morbidity and mortality in elderly patients with sepsis. Initially, immunosenescence was considered as a range of age-related alterations in the immune system. However, increasing evidence has proven that persistent inflammation or even a short-term inflammatory challenge during sepsis could trigger accelerated aging of immune cells, which might further exacerbate inflammatory cytokine storm and promote the shift towards immunosuppression. Thus, premature immunosenescence is found in young sepsis individuals, which further aggravates immune disorders and induces the progression of sepsis. Furthermore, in old sepsis patients, the synergistic effects of both sepsis and aging may cause immunosenescence-associated alterations more significantly, resulting in more severe immune dysfunction and a worse prognosis. Therefore, it is necessary to explore the potential therapeutic strategies targeting immunosenescence during sepsis.
Collapse
Affiliation(s)
- Yanghanzhao Wang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Key laboratory of Perioperative Stress and Protection, Shanghai, China
- Department of Anesthesiology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hao Zhang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China.
- Shanghai Key laboratory of Perioperative Stress and Protection, Shanghai, China.
- Department of Anesthesiology, Shanghai Medical College, Fudan University, Shanghai, China.
| | - Changhong Miao
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China.
- Shanghai Key laboratory of Perioperative Stress and Protection, Shanghai, China.
- Department of Anesthesiology, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
28
|
Li J, Xiang Y, Shentu H, Yang X, Yang J, Ge W, Song E. KLF15 regulates macrophage polarization patterns in deep vein thrombosis. Int Immunopharmacol 2025; 155:114632. [PMID: 40215780 DOI: 10.1016/j.intimp.2025.114632] [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: 01/13/2025] [Revised: 03/27/2025] [Accepted: 04/06/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND KLF15 is involved in cardiovascular disease processes by regulating vascular remodeling and metabolic disorders. Macrophages mediate the inflammatory response in deep vein thrombosis (DVT) by secreting inflammatory cytokines and modulating the fibrinolytic system. Therefore, this study aims to discuss the effect of KLF15 on macrophage polarization in DVT. METHODS In vivo, a DVT animal model was used to assess KLF15 expression and macrophage polarization. In vitro, PMA-treated THP-1 cells with KLF15 overexpression were differentiated into M1- and M2-like macrophages, with polarization markers analyzed by molecular and cellular assays. RESULTS In vivo experiments, levels of KLF15, iNOS, CD206, IL-1β, IL-6, IL-10 and TGF-β were increased in the DVT animal model. In vitro experiments, KLF15 overexpression augmented iNOS, CD86, IL-12, TNF-α, IL-1β, and IL-6 levels in M1-like macrophages. Additionally, KLF15 overexpression diminished CD206, IL-10, ARG1, IL-10 and TGF-β levels in M2-like macrophages. In CUT&Tag, peaks bound to KLF15 and lgG were mainly located in the promoter and intronic regions, and KLF15 protein bound more peaks than lgG near the TSS site. YY1, EIF4E, LCK, HMGB1, GPD2, MORF4L1, HIPK2 and NEK2 were hub genes in the peaks that bind to KLF15. ChIP assay confirmed that KLF15 bound the NEK2 promoter in M1-like macrophages and enhanced its transcription and NF-κB pathway activity. CONCLUSION KLF15 facilitates M1 macrophage polarization in DVT via the NEK2/NF-κB pathway, highlighting its potential as a therapeutic target for DVT management. Future studies are warranted to explore its clinical applicability and mechanistic nuances.
Collapse
Affiliation(s)
- Jizheng Li
- The First Clinical Medical College, The Yunnan University of Chinese Medicine, No.1076 Yuhua Road, Kunming 650500, PR China
| | - YaoYu Xiang
- Dep. of Sports Medicine, The First Affiliated Hospital of Kunming Medical University, No.295 Xichang Road, Kunming 650032, PR China
| | - Haopeng Shentu
- Dep. of Sports Medicine, The First Affiliated Hospital of Kunming Medical University, No.295 Xichang Road, Kunming 650032, PR China
| | - Xianguang Yang
- Dep. of Sports Medicine, The First Affiliated Hospital of Kunming Medical University, No.295 Xichang Road, Kunming 650032, PR China
| | - Jing Yang
- Dep. of Sports Medicine, The First Affiliated Hospital of Kunming Medical University, No.295 Xichang Road, Kunming 650032, PR China
| | - Weiqing Ge
- Dep. of Sports Medicine, The First Affiliated Hospital of Kunming Medical University, No.295 Xichang Road, Kunming 650032, PR China
| | - En Song
- Dep. of Sports Medicine, The First Affiliated Hospital of Kunming Medical University, No.295 Xichang Road, Kunming 650032, PR China.
| |
Collapse
|
29
|
Shi X, Shen T, Gu M, Guan Y, Aimaiti G, Yu W, Zhang X, Yuan WE, Su J. Development of a novel Cu-Mn hydroxide layered nanosheet-loaded drug modulating the tumour microenvironment and enhancing antitumor effects. J Colloid Interface Sci 2025; 696:137904. [PMID: 40393131 DOI: 10.1016/j.jcis.2025.137904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 05/13/2025] [Accepted: 05/15/2025] [Indexed: 05/22/2025]
Abstract
The tumor microenvironment (TME) impedes the effectiveness of therapeutic strategies such as chemodynamic therapy (CDT). This study presents a novel nanoscale drug delivery system designed for the precise release of the chemotherapeutic agent doxorubicin (DOX), aiming to overcome treatment limitations, reduce systemic toxicity, and enhance antitumor efficacy. Mn(III) serves as an immunomodulatory agent, while Cu(II) regulates the levels of glutathione (GSH). Layered double hydroxides (LDHs) were synthesized and efficiently loaded with DOX, followed by surface modification with hyaluronic acid (HA). The HA-coated LDH/DOX nanocarriers showed effective internalization by tumor cells and provided a pH-responsive release of DOX. In vitro, the LDH/HA/DOX complex exhibited strong catalytic activity in the Fenton reaction. In vivo studies using an H22 hepatocarcinoma model confirmed its potent antitumor activity and excellent biocompatibility. Immunohistochemical analyses revealed that treatment with LDH/HA/DOX significantly increased infiltration of M1-polarized tumor-associated macrophages (TAMs), CD4 + T cells and CD8 + T cells, while decreasing M2-polarized TAMs. This change in immune cell profile was associated with notable tumor growth inhibition.
Collapse
Affiliation(s)
- Xiaoying Shi
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China; Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, and School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China; National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai 200240, China; Inner Mongolia Research Institute of Shanghai Jiao Tong University, China
| | - Tianyi Shen
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China; Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, and School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China; National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai 200240, China; Inner Mongolia Research Institute of Shanghai Jiao Tong University, China
| | - Muge Gu
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China; Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, and School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China; National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai 200240, China; Inner Mongolia Research Institute of Shanghai Jiao Tong University, China
| | - Yuanye Guan
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China; Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, and School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China; National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai 200240, China; Inner Mongolia Research Institute of Shanghai Jiao Tong University, China
| | - Gulizeba Aimaiti
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China; Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, and School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China; National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai 200240, China; Inner Mongolia Research Institute of Shanghai Jiao Tong University, China
| | - Wei Yu
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China; Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, and School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China; National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai 200240, China; Inner Mongolia Research Institute of Shanghai Jiao Tong University, China
| | - Xiangqi Zhang
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China; Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, and School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China; National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai 200240, China; Inner Mongolia Research Institute of Shanghai Jiao Tong University, China
| | - Wei-En Yuan
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China; Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, and School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China; National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai 200240, China; Inner Mongolia Research Institute of Shanghai Jiao Tong University, China.
| | - Jing Su
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China; Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, and School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China; National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai 200240, China; Inner Mongolia Research Institute of Shanghai Jiao Tong University, China.
| |
Collapse
|
30
|
Zheng Q, Deng S, Chen X, Wang Y, Yang Y. Macrophage inhibition in the alleviation of nonalcoholic steatohepatitis caused by bariatric surgery. Genes Immun 2025:10.1038/s41435-025-00334-6. [PMID: 40374920 DOI: 10.1038/s41435-025-00334-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 04/21/2025] [Accepted: 05/02/2025] [Indexed: 05/18/2025]
Abstract
The incidence of nonalcoholic steatohepatitis (NASH) is increasing worldwide, and effective treatment is urgently needed. To understand the molecular mechanisms behind the effectiveness of bariatric surgery in treating NASH, we integrated single-cell and bulk RNA sequencing data to identify the role of liver macrophage polarization in alleviating NASH and screen possible drugs for treatment. Analysis revealed that bariatric surgery alleviates NASH by inhibiting liver M1 macrophage polarization with 12 differentially expressed M1 macrophage-related genes. Additionally, 56 potentially effective drugs were predicted for NASH treatment. These findings shed light on the effectiveness of bariatric surgery in treating NASH and offer potential drug candidates for further exploration.
Collapse
Affiliation(s)
- Qianwen Zheng
- Department of Hepatobiliary Surgery, Xi-Jing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| | - Shizhou Deng
- Department of Hepatobiliary Surgery, Xi-Jing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| | - Xiyu Chen
- Department of Hepatobiliary Surgery, Xi-Jing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| | - Yayun Wang
- Specific Lab for Mitochondrial Plasticity Underlying Nervous System Diseases, National Demonstration Center for Experimental Preclinical Medicine Education, The Fourth Military Medical University, Xi'an, 710032, China.
| | - Yanling Yang
- Department of Hepatobiliary Surgery, Xi-Jing Hospital, The Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
31
|
Deng H, Xiao Q, Xu X, Zhang L, Zhang Y. Quercetin Inhibits Gastric Cancer Progression via FAM198B/MAPK Pathway Modulation. Pharmgenomics Pers Med 2025; 18:115-141. [PMID: 40390771 PMCID: PMC12087595 DOI: 10.2147/pgpm.s511324] [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: 01/03/2025] [Accepted: 05/07/2025] [Indexed: 05/21/2025] Open
Abstract
Background The family with the sequence similarity 198 member B (FAM198B) has been found to contribute to the progression of gastric cancer (GC). However, the role and molecular mechanism of FAM198B in GC remains poorly understood. This work found a link between FAM198B and quercetin, and the regulatory effect of FAM198B on the MAPK pathway of GC. Methods FAM198B expression was identified through multiple public data sets and verified in clinical tissue samples. The associations between FAM198B and the prognosis of patients with GC were analyzed via the Kaplan‒Meier plotter and Cox regression analysis. Gene set enrichment analysis, coexpressed genes, and RNA sequencing were used to explore the related functions and signaling pathways of FAM198B in GC. In vitro assays assessed the effects of FAM198B knockdown on GC cells. FAM198B was found as a quercetin target by the HERB database and in vitro assays. Results FAM198B was highly expressed in tissues from GC patients (p<0.001) and was positively associated with poor prognosis (p<0.001) and immune cell infiltration in GC patients. FAM198B knockdown inhibited the proliferation, migration, invasion, and epithelial-mesenchymal transition (EMT) of GC cells (all p<0.05). In addition, FAM198B knockdown decreased the phosphorylation of p-Erk1/2 and p-p38 in GC cells (all p<0.01). Quercetin inhibited FAM198B expression and the phosphorylation of p-Erk1/2 and p-p38 in GC cells (all p<0.05). Conclusion Quercetin inhibits the proliferation, migration, invasion, and EMT of GC cells by inhibiting the FAM198B/MAPK signaling pathway. These discoveries lay the groundwork for developing the treatment of GC by quercetin and targeting FAM198B. In the future, more preclinical and clinical studies are needed to confirm the efficacy and safety of quercetin and target FAM198B in GC.
Collapse
Affiliation(s)
- Hongyang Deng
- Department of General Surgery, Hepatic-Biliary-Pancreatic Institute, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, Gansu, People’s Republic of China
| | - Qi Xiao
- Department of General Surgery, Hepatic-Biliary-Pancreatic Institute, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, Gansu, People’s Republic of China
| | - Xiaodong Xu
- Department of General Surgery, Hepatic-Biliary-Pancreatic Institute, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, Gansu, People’s Republic of China
| | - Lingyi Zhang
- Department of Liver Disease, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, Gansu, People’s Republic of China
| | - Youcheng Zhang
- Department of General Surgery, Hepatic-Biliary-Pancreatic Institute, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, Gansu, People’s Republic of China
| |
Collapse
|
32
|
Fang X, Huang X, Liu W, Lv W, Ying Y, Huang J. Overexpression of FMOD in Thyroid Carcinoma Triggers M1-Like Tumor-Associated Macrophage Polarization by Targeting Rap1B. FASEB J 2025; 39:e70581. [PMID: 40297936 DOI: 10.1096/fj.202403290rr] [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: 12/18/2024] [Revised: 04/14/2025] [Accepted: 04/18/2025] [Indexed: 04/30/2025]
Abstract
Thyroid carcinoma, with limited efficacy of current treatment, influences the lives and health of many people. It is important to explore potential therapeutic targets for thyroid carcinoma. Fibromodulin (FMOD) has been indicated to be connected with the progression of different kinds of tumors, with unknown functions in thyroid carcinoma. In this study, the potential candidate therapeutic targets for thyroid carcinoma were identified by bioinformatics methods, and FMOD was screened out for verification. Cell counting kit-8, wound healing, transwell, and flow cytometry assays were conducted to determine the role of FMOD overexpression in cell viability, migration, invasion, and apoptotic rate of thyroid carcinoma cells, respectively. Subcutaneous tumor growth was monitored in nude mice. Tumor-associated macrophages (TAMs) were co-cultured with thyroid carcinoma cells, and the surface marker of M1-like TAMs, CD80, was identified by flow cytometry. Ras-association proximate 1B (Rap1B), the downstream target of FMOD, was predicted by bioinformatic techniques and validated by Rap1B overexpression rescue. FMOD was identified as a tumor suppressor gene in thyroid carcinoma through bioinformatic techniques. FMOD overexpression inhibited cell viability, migration, and invasion and stimulated apoptosis of thyroid carcinoma cells. In vivo, FMOD upregulation could suppress the growth of solid tumors. Moreover, FMOD overexpression in thyroid carcinoma cells promoted M1-like TAM polarization. FMOD downregulated Rap1B expression in thyroid carcinoma cells, and Rap1B overexpression rescue reversed the impact of FMOD on tumor progression and TAM polarization. In conclusion, FMOD exhibited an inhibitory effect on thyroid carcinoma by stimulating M1-like TAM polarization via targeting Rap1B.
Collapse
Affiliation(s)
- Xiangnan Fang
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Xuemei Huang
- Department of Endocrinology and Metabolism, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Wenfeng Liu
- Department of Endocrinology and Metabolism, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Weiming Lv
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Yong Ying
- Department of Thyroid Hernia Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Jinchang Huang
- Department of Pathology, Ganzhou People's Hospital, Ganzhou, Jiangxi, China
| |
Collapse
|
33
|
Mengru Z, Qinyi W, Zimo Y, Bingqing G, Zhongyu X, Xu J. MXenes in the application of diabetic foot: mechanisms, therapeutic implications and future perspectives. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2025; 36:42. [PMID: 40374863 PMCID: PMC12081522 DOI: 10.1007/s10856-025-06895-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 04/22/2025] [Indexed: 05/18/2025]
Abstract
Diabetic foot represents a significant healthcare challenge, accounting for a substantial portion of diabetes-related hospitalizations and amputations globally. The complexity of diabetic foot management stems from the interplay of poor glycemic control, neuropathy, and peripheral vascular disease, which hinder wound healing processes. The high incidence, recurrence, and amputation rates associated with diabetic foot underscore the urgency for innovative treatment strategies. Recent advancements in nanotechnology, particularly the emergence of MXenes (two-dimensional transition metal carbides and/or nitrides), have shown promising potential in addressing these challenges by offering unique physicochemical and biological properties suitable for various biomedical applications. It is a novel potential strategy for diabetic foot wound healing in the future. This review comprehensively summarizes current knowledge, unique characteristics, and underlying mechanisms of MXenes in the context of diabetic foot management. Additionally, we propose the potential application of MXenes-based therapeutic strategies in diabetes foot. Furthermore, we also provide an overview of their current challenges and the future perspectives in related fields of diabetic wound healing.
Collapse
Affiliation(s)
- Zhang Mengru
- Department of Orthopaedics, Changzhou Hospital Affiliated to Nanjing University of Chinese Medicine, Changzhou, Jiangsu Province, China
| | - Wu Qinyi
- Department of Orthopaedics, Changzhou Hospital Affiliated to Nanjing University of Chinese Medicine, Changzhou, Jiangsu Province, China
| | - Yao Zimo
- The Fourth Clinical School of Nanjing Medical University, Nanjing City, Jiangsu Province, China
| | - Guo Bingqing
- Department of Orthopaedics, Changzhou Hospital Affiliated to Nanjing University of Chinese Medicine, Changzhou, Jiangsu Province, China
| | - Xia Zhongyu
- Department of Orthopaedics, Changzhou Hospital Affiliated to Nanjing University of Chinese Medicine, Changzhou, Jiangsu Province, China.
| | - Jianda Xu
- Department of Orthopaedics, Changzhou Hospital Affiliated to Nanjing University of Chinese Medicine, Changzhou, Jiangsu Province, China.
| |
Collapse
|
34
|
Jin Z, Wei Y, Zhou Z, Fan Z, Huang Y, Liu D. Mechanistic Insights into Maltol-Mediated Reversal of Postmenopausal Osteoporosis via Regulation of CDK14 Ubiquitination in Macrophages. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:11730-11755. [PMID: 40315161 DOI: 10.1021/acs.jafc.5c00545] [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: 05/04/2025]
Abstract
Maltol, primarily derived from Korean red ginseng, exhibits anti-inflammatory properties by modulating macrophage polarization and has potential therapeutic effects on postmenopausal osteoporosis, a condition linked to inflammation. This study explored the molecular mechanisms underlying maltol's ability to inhibit M1 macrophage polarization and regulate osteoblast differentiation via macrophage-mediated pathways. Using in vitro and in vivo models, we demonstrated that maltol upregulates RNF213, which inhibits the CDK14-Pdgfrβ signaling pathway, suppressing M1 polarization and reducing NFκB phosphorylation and pro-inflammatory cytokine production. Additionally, maltol decreases TNFSF12 secretion, mitigating estrogen deficiency-induced osteoblast apoptosis and promoting differentiation. These findings highlight maltol's potential in managing postmenopausal osteoporosis and other inflammatory diseases.
Collapse
Affiliation(s)
- Zhuoru Jin
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China
| | - Yufei Wei
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning 110122, China
| | - Zimo Zhou
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China
| | - Zheng Fan
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China
| | - Ying Huang
- Department of Ultrasound, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China
| | - Da Liu
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China
| |
Collapse
|
35
|
Wu L, Xu H, Zhang X, Zhang M, Xu Y, Zhang Q, Tao H, Dong C, Zhang X, Zhou M, Yang J, Lin C, Song Q. Integrated proteomics and phosphoproteomics profiling dynamic signaling networks underlying two distinct types of macrophage activation. Cell Immunol 2025; 413:104972. [PMID: 40398355 DOI: 10.1016/j.cellimm.2025.104972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Revised: 04/21/2025] [Accepted: 05/11/2025] [Indexed: 05/23/2025]
Abstract
Macrophages play a crucial role in antimicrobial host defense and those with differential maturation/differentiation status differ in inflammatory responses. Herein, GM-CSF and M-CSF primed mouse bone marrow derived macrophages (GM-BMDMs, GM and M-BMDMs, M), the well-established macrophage models in vitro, were utilized and their dynamic signaling changes in response to gram-negative bacteria component LPS treatment were analyzed using both 4D label-free proteomics and phosphoproteomics. Protein changes maintained relatively constant within or across GM and M macrophages post LPS challenge while phospho-protein exhibited more diverse and transient changes. Early induction of phospho-mediated GTPase activities, mRNA processing, and protein-mediated metabolic changes like oxidative phosphorylation (OXPHOS)/mitochondria function was identified at 1 h and maintained until 6 h post LPS treatment in GM and M while canonical TLR mediated MyD88-dependent and -independent pathways were activated at 3 and 6 h, individually at protein levels. Classical and novel phospho-sites for MyD88 and TRIF signaling pathways were also detected by phosphoproteomics. Comprehensively, the integrated protein and phospho-protein trend analysis was conducted and the core protein-phospho-protein network for the early phase actin reorganization, phagocytosis, and TLR signaling in both GM and M were presented. Taken together, these data described differences and similarities between these two types of macrophages in terms of their inflammatory responses to LPS.
Collapse
Affiliation(s)
- Lihong Wu
- Key Laboratory of Marine Drugs, Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Hang Xu
- Key Laboratory of Marine Drugs, Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Xiaonan Zhang
- Innovation Platform of Marine Drug Screening & Evaluation, Qingdao Marine Science and Technology Center, Qingdao 266100, China; Marine Biomedical Research Institute of Qingdao, Qingdao 266071, China
| | - Minghui Zhang
- Key Laboratory of Marine Drugs, Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Yuting Xu
- Key Laboratory of Marine Drugs, Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Qianyue Zhang
- Key Laboratory of Marine Drugs, Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Huiying Tao
- The 2nd Medical College of Binzhou Medical University, Yantai, Shandong 264003, China
| | - Changming Dong
- Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong 264000, China
| | - Xinxin Zhang
- Innovation Platform of Marine Drug Screening & Evaluation, Qingdao Marine Science and Technology Center, Qingdao 266100, China; Marine Biomedical Research Institute of Qingdao, Qingdao 266071, China
| | - Mingming Zhou
- Innovation Platform of Marine Drug Screening & Evaluation, Qingdao Marine Science and Technology Center, Qingdao 266100, China; Marine Biomedical Research Institute of Qingdao, Qingdao 266071, China
| | - Jinbo Yang
- Key Laboratory of Marine Drugs, Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Chunhua Lin
- Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong 264000, China.
| | - Qiaoling Song
- Key Laboratory of Marine Drugs, Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; Innovation Platform of Marine Drug Screening & Evaluation, Qingdao Marine Science and Technology Center, Qingdao 266100, China; Marine Biomedical Research Institute of Qingdao, Qingdao 266071, China.
| |
Collapse
|
36
|
Feng R, Tian F, Zhou J, Ping Y, Han W, Shi X, Bai X, Sun Y, Zhao J, Wu X, Li B. A preliminary study on the promotion of wound healing by paeoniflorin carbon dots loaded in chitosan hydrogel. Biomed Mater 2025; 20:035032. [PMID: 40306299 DOI: 10.1088/1748-605x/add2ba] [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: 12/22/2024] [Accepted: 04/30/2025] [Indexed: 05/02/2025]
Abstract
Due to poor angiogenesis under the wound bed, wound treatment remains a clinical challenge. Therefore, there is an urgent need for new dressings to combat bacterial infections, accelerate angiogenesis, and accelerate wound healing. In this study, we prepared carbon dots nanomaterial (PF-CDs) derived from traditional Chinese medicine paeoniflorin using a simple green one pot hydrothermal method. The average particle size of the CSs we prepared was 4 nm, and a concentration of 200 μg ml-1was ultimately selected for experiments. Subsequently, PF-CDs were loaded into the chitosan hydrogel to form a new type of wound dressing CSMA@PF-CDs hydrogel. CSMA@PF-CDs demonstrated positive biocompatibility by promoting a 20% increase in cell proliferation and strong antibacterial activity. In comparison to the control group, CSMA@PF-CDs enhanced the expression level of anti-inflammatory factors by at least 2.5 times and reduces the expression level of pro-inflammatory factors by at least 3 times. Furthermore, CSMA@PF-CDs promoted the migration of Human umbilical vein endothelial cells and increased vascular endothelial growth factor expression by 5 times. The results ofin vivoexperiments indicate that CSMA@PF-CDs significantly promoted the healing of back wounds in rats. These characteristics make it a promising material for repairing infected wounds and a potential candidate for clinical skin regeneration applications.
Collapse
Affiliation(s)
- Ruiming Feng
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan 030001, Shanxi, People's Republic of China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan 030001, Shanxi, People's Republic of China
| | - Feng Tian
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan 030001, Shanxi, People's Republic of China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan 030001, Shanxi, People's Republic of China
| | - Jian Zhou
- Laboratory for Oral and General Health Integration and Translation, Beijing Tian tan Hospital, Capital Medical University, Beijing 100069, People's Republic of China
| | - Yilin Ping
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan 030001, Shanxi, People's Republic of China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan 030001, Shanxi, People's Republic of China
| | - Wenze Han
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan 030001, Shanxi, People's Republic of China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan 030001, Shanxi, People's Republic of China
| | - Xuexue Shi
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan 030001, Shanxi, People's Republic of China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan 030001, Shanxi, People's Republic of China
| | - Xue Bai
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan 030001, Shanxi, People's Republic of China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan 030001, Shanxi, People's Republic of China
| | - Yufeng Sun
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan 030001, Shanxi, People's Republic of China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan 030001, Shanxi, People's Republic of China
| | - Jiali Zhao
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan 030001, Shanxi, People's Republic of China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan 030001, Shanxi, People's Republic of China
| | - Xiuping Wu
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan 030001, Shanxi, People's Republic of China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan 030001, Shanxi, People's Republic of China
| | - Bing Li
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan 030001, Shanxi, People's Republic of China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan 030001, Shanxi, People's Republic of China
| |
Collapse
|
37
|
Hu HJ, Fu YY, Du SL, Zhang YH, Zhang ZQ, Han GZ. Role of macrophage ATP metabolism disorder in SiO 2‑induced pulmonary fibrosis: a review. Purinergic Signal 2025:10.1007/s11302-025-10093-8. [PMID: 40358809 DOI: 10.1007/s11302-025-10093-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Accepted: 04/30/2025] [Indexed: 05/15/2025] Open
Abstract
Silicosis, a chronic lung disease, results from prolonged inhalation of silica dust (SiO2) in occupational environments, and its pathogenesis remains incompletely elucidated. Studies have shown that alveolar macrophages (AMs) play a pivotal role in its development. These AMs phagocytose the inhaled SiO2, which leads to morphological, structural, and functional abnormalities that result in lung fibrosis. During this process, adenosine triphosphate (ATP) not only provides energy for the physiological and pathological activities but also acts as a key intracellular and extracellular signaling molecule and regulates cytokine synthesis and secretion. This complex process has not been systematically summarized. In this study, first, the current data on ATP metabolism in the development of SiO2-induced pulmonary fibrosis are introduced. ATP metabolism disorder, caused by impaired production, utilization, or distribution of ATP, disrupts macrophage energy homeostasis. Then, how ATP metabolism disorder affects macrophage morphology and function and the inflammatory and fibrotic processes of the lungs by activating the P2X7 receptor-mediated ATP signaling pathway are discussed. Finally, current therapeutic strategies targeting ATP metabolism disorder and ATP signaling pathways in silicosis are summarized. In conclusion, SiO2-induced ATP metabolism disorder indirectly accelerates the progression of silicosis fibrosis.
Collapse
Affiliation(s)
- Hui-Jie Hu
- School of Public Health, Shandong Second Medical University, Weifang, China
- School of Public Health, Jining Medical University, Jining, Shandong, China
| | - Yuan-Yuan Fu
- School of Public Health, Jining Medical University, Jining, Shandong, China
| | - Shu-Ling Du
- School of Public Health, Shandong Second Medical University, Weifang, China
- School of Public Health, Jining Medical University, Jining, Shandong, China
| | - Yu-Han Zhang
- School of Public Health, Jining Medical University, Jining, Shandong, China
| | - Zhao-Qiang Zhang
- School of Public Health, Jining Medical University, Jining, Shandong, China.
| | - Gui-Zhi Han
- School of Public Health, Jining Medical University, Jining, Shandong, China.
| |
Collapse
|
38
|
Feng W, Yang K, Zou Y, Xiao Z, Qian R, Qian R. Progress of ursolic acid on the regulation of macrophage: summary and prospect. Front Immunol 2025; 16:1576771. [PMID: 40421013 PMCID: PMC12104263 DOI: 10.3389/fimmu.2025.1576771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Accepted: 04/15/2025] [Indexed: 05/28/2025] Open
Abstract
Ursolic acid (UA), a prevalent pentacyclic triterpenoid found in numerous fruits and herbs, has garnered significant attention for its vital role in anti-inflammatory processes and immune regulation. The study of immune cells has consistently been a focal point, particularly regarding macrophages, which play crucial roles in antigen presentation, immunomodulation, the inflammatory response, and pathogen phagocytosis. This paper reveals the underlying regulatory effects of UA on the function of macrophages and the specific therapeutic effects of UA on a variety of diseases. Owing to the superior effect of UA on macrophages, different types of macrophages in different tissues have been described. Through the multifaceted regulation of macrophage function, UA may provide new ideas for the development of novel anti-inflammatory and immunomodulatory drugs. However, to facilitate its translation into actual medical means, the specific mechanism of UA in macrophages and its clinical application still need to be further studied.
Collapse
Affiliation(s)
- Wenjing Feng
- Key Laboratory of Vascular Biology and Translational Medicine of Hunan Province, Medical School, Hunan University of Chinese Medicine, Changsha, China
| | - Kehong Yang
- Key Laboratory of Vascular Biology and Translational Medicine of Hunan Province, Medical School, Hunan University of Chinese Medicine, Changsha, China
| | - Ying Zou
- Department of Anatomy, Anatomy Teaching Center of Hunan University of Chinese Medicine, Changsha, China
| | - Zhaohua Xiao
- Xiangya Hospital, Central South University, Changsha, China
| | - Rongkang Qian
- Department of Integrated Traditional Chinese and Western Medicine, Qian Rongkang Clinic, Loudi, China
| | - Ronghua Qian
- Key Laboratory of Vascular Biology and Translational Medicine of Hunan Province, Medical School, Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
39
|
Ji T, Jiang J, Wang X, Yang K, Wang S, Pan B. Single-cell transcriptomics and machine learning unveil ferroptosis features in tumor-associated macrophages: Prognostic model and therapeutic strategies for lung adenocarcinoma. Front Pharmacol 2025; 16:1598756. [PMID: 40421217 PMCID: PMC12104069 DOI: 10.3389/fphar.2025.1598756] [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: 03/24/2025] [Accepted: 04/28/2025] [Indexed: 05/28/2025] Open
Abstract
Background Lung adenocarcinoma (LUAD) is a major cause of cancer-related mortality worldwide. Tumor-associated macrophages (TAMs) play a crucial role in the tumor microenvironment (TME), influencing tumor progression and immune response. Ferroptosis, an iron-dependent form of regulated cell death, has been implicated in tumor biology, but its role within TAMs in LUAD remains unclear. Aim This study aimed to screen key genes associated with ferroptosis in macrophages and construct a prognostic risk model for LUAD based on these genes. Methods Integrating the TCGA-LUAD, GSE131907, and GSE13213 datasets, macrophage heterogeneity was analyzed through single-cell dimensionality reduction clustering, pseudotime analysis, and cell-cell communication. Using the GeneCards ferroptosis gene set (1515 genes), ferroptosis-related differentially expressed genes in macrophages were screened. Eight machine learning algorithms (LASSO, SVM, XGBoost, etc.) were leveraged to identify prognostic genes and build a Cox regression risk model. The functional roles of key genes were validated through immune infiltration analysis, drug sensitivity prediction, and Western blot analysis. Results Single-cell analysis revealed that macrophages in LUAD lead intercellular communication through the MIF (CD74+CXCR4) ligand-receptor interaction, with ferroptosis-related genes (FRGs) highly expressed in macrophages. 73 macrophage FRGs were identified, and through multi-algorithm cross-validation, HLF, HPCAL1, and NUPR1 were determined as core genes. The risk model (Risk Score = HLF × (-0.153) + HPCAL1 × 0.261 + NUPR1 × (-0.21)) demonstrated robust predictive performance in both the TCGA and GSE13213 cohorts, with 1-, 3-, and 5-year AUC values of 0.756, 0.753, and 0.705. The high-risk group was enriched in tumor progression pathways (like epithelial-mesenchymal transition, cell cycle checkpoints), exhibited low expression of immune checkpoint genes (BTLA, CD47), and showed increased sensitivity to cyclophosphamide and crizotinib. Western blotting confirmed the expression levels of HLF, HPCAL1, and NUPR1 were remarkably lower in LUAD cell lines compared to normal bronchial epithelial cells (P < 0.05). Conclusion The research is the first to build a LUAD prognostic model based on macrophage ferroptosis-related genes (HLF, HPCAL1, NUPR1), revealing the immune microenvironment characteristics and drug sensitivity differences in the high-risk group. These findings provide new strategies for precision therapy targeting ferroptosis in tumor-associated macrophages (TAMs).
Collapse
Affiliation(s)
- Ting Ji
- Department of Key Laboratory of Ningxia Stem Cell and Regenerative Medicine, Institute of Medical Sciences, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
- Department of Pulmonary and Critical Care Medicine, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Juanli Jiang
- Department of Neurology, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
- School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, China
| | - Xin Wang
- School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, China
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Kai Yang
- School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, China
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Shaojin Wang
- Department of Key Laboratory of Ningxia Stem Cell and Regenerative Medicine, Institute of Medical Sciences, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
- Department of Pulmonary and Critical Care Medicine, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Bin Pan
- School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, China
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| |
Collapse
|
40
|
Zhou M, Pei B, Cai P, Yi C, Akanyibah FA, Lyu C, Mao F. Human umbilical cord mesenchymal stem cell-derived exosomes repair IBD by activating the SIRT1-FXR pathway in macrophages. Stem Cell Res Ther 2025; 16:233. [PMID: 40346712 PMCID: PMC12065267 DOI: 10.1186/s13287-025-04365-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Accepted: 04/24/2025] [Indexed: 05/11/2025] Open
Abstract
BACKGROUND Inflammatory bowel disease (IBD), a chronic immune disorder, has increasing global incidence and poor treatment outcome. Abnormal macrophage function is implicated in the pathophysiology of IBD. In this study, we investigated the mechanism by which human umbilical cord mesenchymal stem cell-derived exosomes (hucMSC-Ex) inhibit inflammation in IBD mouse and macrophage inflammation models. METHODS We established a dextran sodium sulfate (DSS)-induce BALB/c mice model of IBD and treated with hucMSC-Ex via tail vein to evaluate their repair effect on IBD mice. An in vitro macrophage inflammation model was established using lipopolysaccharide (LPS) and Nigericin (Nig) by stimulating mouse macrophage RAW264.7 and human myeloid leukemia mononuclear (THP-1) cells to assess the repair effect of hucMSC-Ex on macrophage inflammation. EX 527, an effective inhibitor of silent information regulator of transcription 1 (SIRT1), was employed in both the in vivo and in vitro models to explore the effect of hucMSC-Ex on the SIRT1-FXR (farnesoid X receptor) pathway in macrophages during the attenuation of inflammation. RESULTS HucMSC-Ex effectively inhibited inflammation in both the in vivo and in vitro models by up-regulating the expressions of SIRT1 and FXR, which reduced the acetylation level of FXR and inhibited the activation of NOD-like receptor thermal protein domain associated protein 3 (NLRP3) inflammasome. The addition of EX 527 further proved that hucMSC-Ex can reduce the acetylation of FXR by activating the SIRT1-FXR pathway, and the decrease of FXR acetylation was directly related to the inhibition of the activity of the NLRP3 inflammasome. CONCLUSION HucMSC-Ex alleviates IBD by reducing the acetylation level of FXR through activating the SIRT1-FXR pathway in macrophages and directly negatively regulating the activation of NLRP3 inflammasomes, thus inhibiting the occurrence of the inflammatory process.
Collapse
Affiliation(s)
- Mengjiao Zhou
- Department of Laboratory Medicine, The Affiliated People's Hospital, Jiangsu University, No.8 Dianli Road, Zhenjiang, Jiangsu, 212002, P. R. China
- Institute of Hematology, Jiangsu University, Zhenjiang, Jiangsu, 212013, P. R. China
| | - Bing Pei
- Department of Clinical Laboratory, The Affiliated Suqian First People's Hospital of Nanjing Medical University, Suqian, Jiangsu, 223800, P. R. China
| | - Peipei Cai
- Department of Laboratory Medicine, The Affiliated People's Hospital, Jiangsu University, No.8 Dianli Road, Zhenjiang, Jiangsu, 212002, P. R. China
| | - Chengxue Yi
- School of Medical Technology, Zhenjiang College, Zhenjiang, Jiangsu, 212028, P. R. China
| | - Francis Atim Akanyibah
- Department of Laboratory Medicine, The Affiliated People's Hospital, Jiangsu University, No.8 Dianli Road, Zhenjiang, Jiangsu, 212002, P. R. China
| | - Changkun Lyu
- School of Medical Technology, Shangqiu Medical College Shangqiu, Shangqiu, Henan, 476100, P. R. China
| | - Fei Mao
- Department of Laboratory Medicine, The Affiliated People's Hospital, Jiangsu University, No.8 Dianli Road, Zhenjiang, Jiangsu, 212002, P. R. China.
- Institute of Hematology, Jiangsu University, Zhenjiang, Jiangsu, 212013, P. R. China.
| |
Collapse
|
41
|
Zhou J, Wu K, Ma Y, Zhu J, Zhou Y, Zhang Z, Li F, Zeng G, Li S, Tan S, Zhang Y, Wan C, Tu T, Lin Q, Liu Q. GTS-21 alleviates sepsis-induced atrial fibrillation susceptibility by modulating macrophage polarization and Neuregulin-1 secretion. Int Immunopharmacol 2025; 154:114561. [PMID: 40186903 DOI: 10.1016/j.intimp.2025.114561] [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: 12/15/2024] [Revised: 03/02/2025] [Accepted: 03/23/2025] [Indexed: 04/07/2025]
Abstract
OBJECTIVE Sepsis-induced atrial fibrillation (AF) is driven by systemic inflammation and macrophage-mediated atrial remodeling, with proinflammatory M1 macrophages playing a key role. This study investigates whether GTS-21, an α7nAChR agonist, can reduce AF susceptibility by promoting macrophage polarization towards the anti-inflammatory M2 phenotype. METHODS A mouse model of lipopolysaccharide (LPS) (10 mg/kg)-induced sepsis was used to explore the relationship between atrial inflammation and AF. GTS-21 (20 mg/kg) was administered to assess its impact on 48-h survival and AF incidence. Cardiac function was evaluated using echocardiography. Markers of myocardial injury, including CK-MB, LDH, and cTnI, were measured. Macrophage polarization and atrial inflammation were assessed using immunofluorescence, flow cytometry, RT-qPCR, and western blotting. Oxidative stress and mitochondrial function were evaluated using reactive oxygen species (ROS) measurements, electron microscopy, and mitochondrial protein expression analysis. Calcium dynamics were studied using western blotting and confocal microscopy. RESULTS In LPS-induced septic mice, GTS-21 improved 48-h survival rates and reduced the induction rate and duration of AF (P < 0.05). Echocardiography showed a preserved left ventricular ejection fraction and enhanced diastolic function. Mechanistically, it promoted M2 macrophage polarization, inhibited the NF-κB P65/NLRP3/C-caspase 1 pathway to reduce IL-1β release, and alleviated oxidative stress. Additionally, mitochondrial structure was restored by reversing fission and promoting fusion, while calcium-handling proteins (NCX-1, RYR2, and SERCA2a) were regulated to prevent intracellular calcium overload, reducing AF susceptibility. CONCLUSION GTS-21 mitigated atrial inflammation and reduced the incidence of AF in mice with sepsis by regulating macrophage polarization, reducing oxidative stress, and preserving mitochondrial and calcium dynamics in cardiomyocytes. These findings highlight the therapeutic potential of GTS-21 in treating sepsis-induced AF.
Collapse
Affiliation(s)
- Jiabao Zhou
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, PR China; Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province, Changsha, Hunan 410011, PR China; Cardiovascular Disease Research Center of Hunan Province, Changsha, Hunan 410011, PR China; Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, Hunan 410011, PR China
| | - Keke Wu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, PR China; Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province, Changsha, Hunan 410011, PR China; Cardiovascular Disease Research Center of Hunan Province, Changsha, Hunan 410011, PR China; Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, Hunan 410011, PR China
| | - Yingxu Ma
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, PR China; Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province, Changsha, Hunan 410011, PR China; Cardiovascular Disease Research Center of Hunan Province, Changsha, Hunan 410011, PR China; Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, Hunan 410011, PR China
| | - Jiayi Zhu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, PR China; Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province, Changsha, Hunan 410011, PR China; Cardiovascular Disease Research Center of Hunan Province, Changsha, Hunan 410011, PR China; Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, Hunan 410011, PR China
| | - Yong Zhou
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, PR China; Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province, Changsha, Hunan 410011, PR China; Cardiovascular Disease Research Center of Hunan Province, Changsha, Hunan 410011, PR China; Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, Hunan 410011, PR China
| | - Zixi Zhang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, PR China; Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province, Changsha, Hunan 410011, PR China; Cardiovascular Disease Research Center of Hunan Province, Changsha, Hunan 410011, PR China; Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, Hunan 410011, PR China
| | - Fanqi Li
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, PR China; Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province, Changsha, Hunan 410011, PR China; Cardiovascular Disease Research Center of Hunan Province, Changsha, Hunan 410011, PR China; Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, Hunan 410011, PR China
| | - Gaoming Zeng
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, PR China; Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province, Changsha, Hunan 410011, PR China; Cardiovascular Disease Research Center of Hunan Province, Changsha, Hunan 410011, PR China; Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, Hunan 410011, PR China
| | - Shunyi Li
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, PR China; Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province, Changsha, Hunan 410011, PR China; Cardiovascular Disease Research Center of Hunan Province, Changsha, Hunan 410011, PR China; Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, Hunan 410011, PR China
| | - Siyuan Tan
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, PR China; Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province, Changsha, Hunan 410011, PR China; Cardiovascular Disease Research Center of Hunan Province, Changsha, Hunan 410011, PR China; Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, Hunan 410011, PR China
| | - Yusha Zhang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, PR China; Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province, Changsha, Hunan 410011, PR China; Cardiovascular Disease Research Center of Hunan Province, Changsha, Hunan 410011, PR China; Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, Hunan 410011, PR China
| | - Cancan Wan
- First Clinical College, Changsha Medical University, Changsha, Hunan 410219, PR China
| | - Tao Tu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, PR China; Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province, Changsha, Hunan 410011, PR China; Cardiovascular Disease Research Center of Hunan Province, Changsha, Hunan 410011, PR China; Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, Hunan 410011, PR China
| | - Qiuzhen Lin
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, PR China; Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province, Changsha, Hunan 410011, PR China; Cardiovascular Disease Research Center of Hunan Province, Changsha, Hunan 410011, PR China; Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, Hunan 410011, PR China.
| | - Qiming Liu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, PR China; Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province, Changsha, Hunan 410011, PR China; Cardiovascular Disease Research Center of Hunan Province, Changsha, Hunan 410011, PR China; Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, Hunan 410011, PR China.
| |
Collapse
|
42
|
Fu J, Chen X, Li J, Peng L. Research advances of Sappanone A in inflammation-related diseases. Front Med (Lausanne) 2025; 12:1569732. [PMID: 40406412 PMCID: PMC12095284 DOI: 10.3389/fmed.2025.1569732] [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: 02/01/2025] [Accepted: 04/21/2025] [Indexed: 05/26/2025] Open
Abstract
Sappanone A (SA), a kind of homoisoflavanone extracted from the dry heartwood of Caesalpinia sappan L., has been shown to possess diverse bioactivities involving anti-inflammatory, antioxidant, and anti-apoptotic properties. Sustained proinflammatory state is a major factor in the occurrence and development of various diseases. Given the characteristics of SA, many studies have explored the effect of SA on inflammation-related diseases, which uncovered the multifaceted therapeutic potential of SA in such diseases. In this mini-review, we summarized the current achievements of SA on inflammation-related diseases (such as myocardial ischemia-reperfusion injury, liver injury, respiratory diseases, and kidney injury, etc.), in order to provide useful insights into the role of SA in inflammation-related diseases and benefit future clinical applications.
Collapse
Affiliation(s)
- Jie Fu
- Department of Neurology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Xiu Chen
- Department of Neurology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jinglun Li
- Department of Neurology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Lilei Peng
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
43
|
Tsou TC, Yeh SC, Tsai FY, Chen PY. Palmitic acid and lipopolysaccharide induce macrophage TNFα secretion, suppressing browning regulators and mitochondrial respiration in adipocytes. Toxicol Appl Pharmacol 2025; 500:117389. [PMID: 40348028 DOI: 10.1016/j.taap.2025.117389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 05/04/2025] [Accepted: 05/07/2025] [Indexed: 05/14/2025]
Abstract
Obesity and its associated pro-inflammatory activity contribute significantly to metabolic dysfunction. In contrast, browning of white adipose tissue (WAT) generally improves metabolic health. Our prior research suggested that macrophage-derived pro-inflammatory cytokines suppress key regulators of browning-adrenergic receptor β3 (Adrb3) and peroxisome proliferator-activated receptor γ (Pparg)-as well as energy metabolism mediators-insulin receptor substrate 1 (Irs1) and hormone-sensitive lipase (Lipe)-in diet-induced obese mice. To explore this mechanism, we developed an in vitro model using RAW264.7 macrophages and 3T3-L1 adipocytes exposed to palmitic acid (PA) and/or lipopolysaccharide (LPS). PA (200 μM) and LPS (1.0 μg/ml) synergistically promoted M1 polarization of macrophages and secretion of pro-inflammatory cytokines, with tumor necrosis factor-α (TNFα), C-C motif chemokine ligand 2 (CCL2), CCL5, and interleukin-6 (IL-6) being predominant. Conditioned media from both control and PA-treated macrophages, when exposed to LPS ≥0.01 μg/ml, significantly downregulated Adrb3, Pparg, Irs1, and Lipe in adipocytes. At physiologically relevant LPS levels (≤0.001 μg/ml), PA-treated macrophage media exerted greater suppression of these genes than controls. Among the cytokines, TNFα emerged as the primary mediator, significantly reducing expression of the four key regulators. Furthermore, adipocytes treated with TNFα exhibited significant reductions in both uncoupling protein 1 (Ucp1) expression and mitochondrial respiration. These findings demonstrate that exposure to obesity-associated factors (PA and LPS) induces macrophage-derived TNFα, which suppresses browning and mitochondrial function in adipocytes. This mechanism may inform new therapeutic strategies targeting TNFα to alleviate obesity-related metabolic disorders.
Collapse
Affiliation(s)
- Tsui-Chun Tsou
- National Institute of Environmental Health Sciences, National Health Research Institutes, Zhunan, Miaoli County 35053, Taiwan.
| | - Szu-Ching Yeh
- National Institute of Environmental Health Sciences, National Health Research Institutes, Zhunan, Miaoli County 35053, Taiwan
| | - Feng-Yuan Tsai
- National Institute of Environmental Health Sciences, National Health Research Institutes, Zhunan, Miaoli County 35053, Taiwan
| | - Pei-Yu Chen
- National Institute of Environmental Health Sciences, National Health Research Institutes, Zhunan, Miaoli County 35053, Taiwan
| |
Collapse
|
44
|
Liu M, Zou G, Lu M, Fu J, Chen H, Pan C, Liu HM, Fu L. Mechanism of Rabdosia rubescens extract against gastric cancer microenvironment by SIRT1/NF-κB/p53 pathway and promoting tumor-associated macrophage polarization. JOURNAL OF ETHNOPHARMACOLOGY 2025; 349:119935. [PMID: 40345273 DOI: 10.1016/j.jep.2025.119935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 04/23/2025] [Accepted: 05/06/2025] [Indexed: 05/11/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The traditional action of Rabdosia rubescens (Hemsl.) H. Hara is heat-clearing and detoxifying, relieve sore throat, dissipate binds and disperse swelling. DLC, as an extract prepared from Rabdosiae Rubescentis Herba, could regulate the polarization of tumor associated macrophages (TAMs). For TAMs play an important role in the tumor microenvironment. It is worthy to further explore the mechanism of DLC on the polarized function of macrophages. AIM OF THE STUDY The aim of this study is to investigate the activity and molecular mechanisms of DLC on dissipating binds and dispersing swelling by modulating the gastric cancer microenvironment and macrophage polarization. MATERIALS AND METHODS We conducted comprehensive qualitative and quantitative chromatographic analyses to characterize the main components of DLC. To evaluate its anti-tumor effects, immunofluorescence, MTT assay, plate cloning, transcriptomics analysis, western blotting, and siRNA knockdown experiments were performed to assess DLC's action on gastric cancer cell proliferation. Additionally, we utilized Trypan blue staining, a THP-1 and MGC-803 co-culture model, flow cytometry, enzyme-linked immunosorbent assay (ELISA), and a mouse xenograft model with five distinct dosage groups to systematically investigate DLC's effects on macrophage polarization. RESULTS Key compounds in DLC were identified. The vivo tests demonstrated the tumor inhibition rate of the 5 g/kg DLC group reached 66.99 %, surpassing that of the 5-fluorouracil group (59.94 %). Mechanistically, DLC upregulated SIRT1 expression and suppressed NF-κB pathway, thereby preventing p65 from translocating into nuclear and modulating downstream p53/MDM2/USP7 signaling. Moreover, DLC enhanced M1 macrophage factors such as TNF-α, IL-6 while inhibiting M2 marker TGF-β, effectively repolarizing M2 TAMs toward an M1 phenotype. This effect was associated with suppressed protein expression of HIF-1α, p-p65, and p-PI3K. CONCLUSION This study provides insights into DLC's mechanisms in regulating tumor microenvironment remodeling and promoting macrophage polarization toward an anti-tumor phenotype. These results provide a solid basis for DLC's potential clinical treament in gastric cancer, highlighting its promise as a natural therapeutic agent.
Collapse
Affiliation(s)
- Mengran Liu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou, 450001, China; Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou, 450001, China
| | - Guona Zou
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou, 450001, China; Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou, 450001, China
| | - Mengyao Lu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou, 450001, China; Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou, 450001, China
| | - Jiayue Fu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou, 450001, China; Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou, 450001, China
| | - Han Chen
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou, 450001, China; Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou, 450001, China
| | - Chengxue Pan
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou, 450001, China; Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou, 450001, China
| | - Hong-Min Liu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou, 450001, China; Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou, 450001, China.
| | - Ling Fu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou, 450001, China; Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou, 450001, China.
| |
Collapse
|
45
|
Groves PL, Hockey L, O’Sullivan BJ, Zhang LY, Xiong Z, Nguyen QH, Tan ME, Lutzky VP, Davis RA, Chambers DC, Apte SH. Transcriptomic Plasticity of Human Alveolar Macrophages Revealed by Single-Cell RNA Sequencing Following Drug Exposure: Implications for Therapeutic Development. Int J Mol Sci 2025; 26:4439. [PMID: 40362676 PMCID: PMC12072627 DOI: 10.3390/ijms26094439] [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: 03/15/2025] [Revised: 04/24/2025] [Accepted: 04/29/2025] [Indexed: 05/15/2025] Open
Abstract
Alveolar macrophages (AM) must perform three seemingly opposing roles including homeostasis, driving inflammation, and facilitating tissue repair. Whilst there is now consensus (supported by a large body of human single cell RNA sequencing (scRNA-seq) data) that the cell subsets that perform these tasks can readily be found based on their transcriptome, their ontogeny has remained unclear. Moreover, there is agreement that in all types of pulmonary fibrosis (PF) there is an expanded population of profibrotic AM that may aberrantly drive PF. From a therapeutic viewpoint, there is great appeal in the notion that the transcriptional program in different AM subsets is not fixed but remains plastic and amenable to pharmacological reprogramming. Accordingly, this study addresses this question by performing scRNA-seq on human AM following treatment with drugs or perturbagens including pioglitazone, trametinib, nintedanib, lipopolysaccharide and the natural compound endiandrin A. Each treatment induced a unique global transcriptional change, driving the cells towards distinct subsets, further supported by trajectory analysis, confirming a high level of plasticity. Confirmatory experiments using qPCR demonstrated that single exposure to a compound induced a relatively stable transcriptome, whereas serial exposure to a different compound allowed the cells to be reprogrammed yet again to a different phenotype. These findings add new insight into the biology of AM and support the development of novel therapies to treat PF.
Collapse
Affiliation(s)
- Penny L. Groves
- Queensland Lung Transplant Service, The Prince Charles Hospital, Chermside, QLD 4032, Australia; (P.L.G.); (B.J.O.); (L.-Y.Z.); (V.P.L.); (D.C.C.)
| | - Levi Hockey
- Faculty of Health, Medicine and Behavioural Sciences, The University of Queensland, St. Lucia, QLD 4072, Australia;
- QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; (Z.X.); (Q.H.N.)
| | - Brendan J. O’Sullivan
- Queensland Lung Transplant Service, The Prince Charles Hospital, Chermside, QLD 4032, Australia; (P.L.G.); (B.J.O.); (L.-Y.Z.); (V.P.L.); (D.C.C.)
- Faculty of Health, Medicine and Behavioural Sciences, The University of Queensland, St. Lucia, QLD 4072, Australia;
| | - Lai-Ying Zhang
- Queensland Lung Transplant Service, The Prince Charles Hospital, Chermside, QLD 4032, Australia; (P.L.G.); (B.J.O.); (L.-Y.Z.); (V.P.L.); (D.C.C.)
- Faculty of Health, Medicine and Behavioural Sciences, The University of Queensland, St. Lucia, QLD 4072, Australia;
| | - Zherui Xiong
- QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; (Z.X.); (Q.H.N.)
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD 4072, Australia
| | - Quan H. Nguyen
- QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; (Z.X.); (Q.H.N.)
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD 4072, Australia
| | - Maxine E. Tan
- Queensland Lung Transplant Service, The Prince Charles Hospital, Chermside, QLD 4032, Australia; (P.L.G.); (B.J.O.); (L.-Y.Z.); (V.P.L.); (D.C.C.)
- Faculty of Health, Medicine and Behavioural Sciences, The University of Queensland, St. Lucia, QLD 4072, Australia;
| | - Viviana P. Lutzky
- Queensland Lung Transplant Service, The Prince Charles Hospital, Chermside, QLD 4032, Australia; (P.L.G.); (B.J.O.); (L.-Y.Z.); (V.P.L.); (D.C.C.)
- Faculty of Health, Medicine and Behavioural Sciences, The University of Queensland, St. Lucia, QLD 4072, Australia;
| | - Rohan A. Davis
- School of Environment and Science, Griffith University, Nathan, QLD 4111, Australia;
- NatureBank, Institute for Biomedicine and Glycomics, Griffith University, Nathan, QLD 4111, Australia
| | - Daniel C. Chambers
- Queensland Lung Transplant Service, The Prince Charles Hospital, Chermside, QLD 4032, Australia; (P.L.G.); (B.J.O.); (L.-Y.Z.); (V.P.L.); (D.C.C.)
- Faculty of Health, Medicine and Behavioural Sciences, The University of Queensland, St. Lucia, QLD 4072, Australia;
| | - Simon H. Apte
- Queensland Lung Transplant Service, The Prince Charles Hospital, Chermside, QLD 4032, Australia; (P.L.G.); (B.J.O.); (L.-Y.Z.); (V.P.L.); (D.C.C.)
- Faculty of Health, Medicine and Behavioural Sciences, The University of Queensland, St. Lucia, QLD 4072, Australia;
| |
Collapse
|
46
|
Sun G, Wang Y, Ni K, Shen J, Liu D, Wang H. COL5A2-mediated endoplasmic reticulum stress promotes macrophage M2 polarization in lung adenocarcinoma. Cell Stress Chaperones 2025; 30:100081. [PMID: 40345640 DOI: 10.1016/j.cstres.2025.100081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 04/22/2025] [Accepted: 05/02/2025] [Indexed: 05/11/2025] Open
Abstract
Collagen is a major component of the extracellular matrix. Type V collagen α2 (COL5A2), a common collagen subtype, plays a crucial role in immune regulation, angiogenesis, and tumor metastasis. It is highly expressed in various malignancies, but its mechanistic role in lung adenocarcinoma (LUAD) remains unclear. Therefore, this study aims to investigate the regulatory mechanism of COL5A2 in mediating macrophage M2 polarization in LUAD. We analyzed COL5A2 expression in LUAD samples from the TCGA-LUAD database. Using GSEA, we sought to identify the signaling pathways influenced by COL5A2 expression. mRNA levels of COL5A2, TGF-β, and IL-10 were quantified via qPCR analysis, and protein levels of COL5A2, PD-L1, and endoplasmic reticulum (ER) stress-related proteins (GRP78 and CHOP) were assessed using western blot. Immunofluorescence assay detected the fluorescence signal of CD206 in M2 macrophages, while flow cytometry assessed the M2 macrophage marker CD206, flow cytometry determined the positive rates for CD68 and CD206. Exosome uptake by macrophages was examined using confocal microscopy, and cell viability was measured with cell counting kit-8. KI-67 protein expression was analyzed by immunohistochemistry, and in vivo assays in animals verified our findings. The results showed that elevated COL5A2 levels in LUAD were found to correlate with a shift toward M2 macrophage polarization. Specifically, the overexpression of COL5A2 amplified ER stress, which led to an increase in PD-L1 exosome release and macrophage uptake of PD-L1, thus driving the M2 phenotype. In conclusion, COL5A2 in LUAD induces ER stress, which is associated with elevated PD-L1 exosome secretion and macrophage PD-L1 uptake, ramping up M2 polarization in macrophages.
Collapse
Affiliation(s)
- Gaozhong Sun
- Department of Thoracic Surgery, Cancer Center, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, China
| | - Yanzhe Wang
- The Second School of Clinical Medicine of Zhejiang Chinese Medical University, Hangzhou, China
| | - Kewei Ni
- Department of Thoracic Surgery, Cancer Center, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, China
| | - Jian Shen
- Department of Thoracic Surgery, Cancer Center, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, China
| | - Dongdong Liu
- Department of Thoracic Surgery, Cancer Center, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, China
| | - Haitao Wang
- Department of Thoracic Surgery, Cancer Center, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, China.
| |
Collapse
|
47
|
Liu L, Wuyun T, Sun X, Zhang Y, Cha G, Zhao L. Therapeutic efficacy of TMTP1-modified EVs in overcoming bone metastasis and immune resistance in PIK3CA mutant NSCLC. Cell Death Dis 2025; 16:367. [PMID: 40328748 PMCID: PMC12055990 DOI: 10.1038/s41419-025-07685-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: 10/20/2024] [Revised: 04/08/2025] [Accepted: 04/17/2025] [Indexed: 05/08/2025]
Abstract
Non-small cell lung cancer (NSCLC) with PIK3CA mutations demonstrates significant challenges in treatment due to enhanced bone metastasis and immune checkpoint resistance. This study investigates the efficacy of tumor-targeting peptide 1-modified cancer stem cell-derived extracellular vesicles (TMTP1-TSRP-EVs) in reshaping the tumor microenvironment and reversing immune checkpoint resistance in NSCLC. By integrating TMTP1-TSRP into EVs, we aim to specifically deliver therapeutic agents to NSCLC cells, focusing on inhibiting the PI3K/Akt/mTOR pathway, a crucial driver of oncogenic activity and immune evasion in PIK3CA-mutated cells. Our comprehensive in vitro and in vivo analyses show that TMTP1-TSRP-EVs significantly inhibit tumor growth, reduce PD-L1 expression, and enhance CD8+ T cell infiltration, effectively reversing the immune-suppressive microenvironment. Moreover, the in vivo models confirm that our approach not only suppresses bone metastases but also overcomes primary resistance to immune checkpoint inhibitors by modulating the expression of key immunological markers. These findings suggest that targeted delivery of TMTP1-TSRP-EVs could provide a novel therapeutic strategy for treating PIK3CA-mutant NSCLC, offering significant improvements over traditional therapies by directly targeting the molecular pathogenesis of tumor resistance and metastasis. Molecular Mechanisms Reshaping the TME to Halt PI3K-Mutant Bone Metastasis of NSCLC and Overcoming Primary ICI Resistance. (Created by BioRender).
Collapse
Affiliation(s)
- Liwen Liu
- Department of Radiology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Tanghesi Wuyun
- The Second Department of Respiratory, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xin Sun
- The Second Department of Respiratory, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yu Zhang
- The Second Department of Respiratory, Harbin Medical University Cancer Hospital, Harbin, China
| | - Geqi Cha
- Department of Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Ling Zhao
- The Second Department of Respiratory, Harbin Medical University Cancer Hospital, Harbin, China.
| |
Collapse
|
48
|
Zhao Y, Pei L, Liu B, Mao Z, Niu Y, Li S, Yang M, Liu W, Hai H, Luo Y, Liu T. Macrophage Membrane-Coated Nanomedicine Enhances Cancer Immunotherapy by Activating Macrophages and T Cells. Mol Pharm 2025; 22:2402-2412. [PMID: 40266070 DOI: 10.1021/acs.molpharmaceut.4c00950] [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: 04/24/2025]
Abstract
Cancer immunotherapy has transformed malignancy treatment, but the tumor microenvironment (TME) presents significant obstacles. PD-1 blockade therapy, while widely used, faces issues such as resistance, adverse events, and limited predictive biomarkers. Therefore, novel therapeutic strategies are needed to enhance their efficacy and safety. Tumor-associated macrophages (TAMs), often exhibiting an anti-inflammatory M2 phenotype, contribute to poor prognoses and treatment resistance. Targeting TAMs to repolarize them to a pro-inflammatory M1 state can alleviate immunosuppression and enhance T cell-mediated antitumor responses. TMP195, a class IIa histone deacetylase inhibitor, has shown potential in reprogramming TAMs and synergizing with anti-PD-1 antibodies, although clinical application challenges exist. This study aimed to enhance the PD-1 blockade immunotherapy effectiveness by activating tumor-killing macrophages and T cells using biomimetic nanomedicines. A novel macrophage cell membrane-coated PLGA nanoparticle loaded with small molecule inhibitor, TMP195 (M1@PLGA-PEG-TMP195), was designed, prepared, and characterized. This macrophage membrane-coated PLGA nanoparticle delivery system had good drug loading and cancer cell targeting ability. This approach repolarized TAMs to M1 phenotypes and, combined with PD-1 inhibitors, achieved synergistic cancer treatment effects, improving therapeutic efficacy and inhibiting breast cancer growth and metastasis.
Collapse
Affiliation(s)
- Yongmei Zhao
- School of Pharmacy, Nantong University, Nantong 226019, China
| | - Lulu Pei
- School of Pharmacy, Nantong University, Nantong 226019, China
| | - Baolin Liu
- School of Pharmacy, Nantong University, Nantong 226019, China
| | - Zehao Mao
- School of Pharmacy, Nantong University, Nantong 226019, China
| | - Yingyi Niu
- School of Pharmacy, Nantong University, Nantong 226019, China
| | - Siqi Li
- School of Pharmacy, Nantong University, Nantong 226019, China
| | - Meiqing Yang
- School of Pharmacy, Nantong University, Nantong 226019, China
| | - Wenqian Liu
- School of Pharmacy, Nantong University, Nantong 226019, China
| | - Hongde Hai
- School of Pharmacy, Nantong University, Nantong 226019, China
| | - Yunyao Luo
- School of Pharmacy, Nantong University, Nantong 226019, China
| | - Tianqing Liu
- NICM Health Research Institute, Western Sydney University, Westmead, Sydney 2145, Australia
| |
Collapse
|
49
|
Chen L, Liu Y, Yu C, Cao P, Ma Y, Geng Y, Cai Y, Zhang Y, Liu J, Li Y, Luan Q. Induced pluripotent stem cell-derived mesenchymal stem cells (iMSCs) inhibit M1 macrophage polarization and reduce alveolar bone loss associated with periodontitis. Stem Cell Res Ther 2025; 16:223. [PMID: 40317064 PMCID: PMC12046914 DOI: 10.1186/s13287-025-04327-0] [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: 12/06/2024] [Accepted: 04/09/2025] [Indexed: 05/04/2025] Open
Abstract
BACKGROUND Periodontitis is a chronic inflammatory disease and macrophages play a pivotal role in the progression of periodontitis. Mesenchymal stem cells (MSCs) have emerged as potential therapeutic agents for the treatment of periodontitis due to their immunomodulatory properties and capacity for tissue regeneration. Compared to conventionally derived MSCs, induced pluripotent stem cell-derived MSCs (iMSCs) offer distinct advantages as promising candidates for MSC-based therapies, owing to their non-invasive acquisition methods and virtually unlimited availability. This study aims to investigate the effects and mechanisms of iMSCs in modulating macrophage polarization and alleviating periodontitis-related alveolar bone loss. METHODS iMSCs were generated from iPSCs and characterized for differentiation potential. The effects of iMSCs on macrophage polarization were evaluated using THP-1-derived macrophages under inflammatory conditions (LPS and IFN-γ stimulation). Co-culture assays, cytokine analysis, reactive oxygen species (ROS) detection, transcriptomic analysis, flow cytometry, reverse transcription-quantitative polymerase chain reaction (RT-qPCR), and western blot analysis were performed to elucidate the underlying mechanisms. The therapeutic potential of iMSCs was assessed in a ligature-induced periodontitis mouse model using micro-CT, histological analysis, and immunofluorescence staining. RESULTS iMSCs inhibit M1 macrophage polarization through the suppression of the NF-κB signaling pathway. Additionally, iMSCs reduce the production of pro-inflammatory cytokines (IL-1β, IL-17) and reactive oxygen species (ROS), while enhancing the secretion of anti-inflammatory cytokines (IL-10) and growth factors (VEGF), thereby improving the inflammatory microenvironment. Under inflammatory conditions, iMSCs preserve the osteogenic potential of periodontal ligament stem cells (PDLSCs) and alleviate alveolar bone loss in mice with periodontitis. In vivo, iMSCs reduce the number of M1 macrophages and inhibit the activation of NF-κB in periodontal tissues, supporting their anti-inflammatory and immunomodulatory effects. CONCLUSION iMSCs demonstrate significant therapeutic potential in periodontitis by modulating macrophage polarization, reducing oxidative stress, and mitigating alveolar bone loss associated with the disease. These findings provide new insights into the mechanisms of iMSCs and their application as cell-based therapies for periodontal diseases.
Collapse
Affiliation(s)
- Liang Chen
- Department of Periodontology, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, No. 22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, People's Republic of China
| | - Yuanqing Liu
- Department of Periodontology, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, No. 22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, People's Republic of China
| | - Chenhao Yu
- Department of Periodontology, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, No. 22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, People's Republic of China
| | - Pei Cao
- Department of Periodontology, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, No. 22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, People's Republic of China
| | - Yiming Ma
- Department of Periodontology, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, No. 22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, People's Republic of China
| | - Yiran Geng
- Department of Periodontology, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, No. 22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, People's Republic of China
| | - Yu Cai
- Department of Periodontology, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, No. 22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, People's Republic of China
| | - Yong Zhang
- First Clinical Division, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, No. 22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, People's Republic of China
| | - Jia Liu
- Department of Periodontology, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, No. 22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, People's Republic of China
| | - Yang Li
- Department of Cell Biology, School of Basic Medical Sciences, Peking University Stem Cell Research Center, Peking University, Beijing, People's Republic of China.
| | - Qingxian Luan
- Department of Periodontology, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, No. 22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, People's Republic of China.
| |
Collapse
|
50
|
Cao M, Deng Y, Hao Q, Yan H, Wang QL, Dong C, Wu J, He Y, Huang LB, Xia X, Gao Y, Chen HN, Zhang WH, Zhang YJ, Zhuo X, Dai L, Hu H, Peng Y, Zhang F, Liu Z, Huang W, Zhang H, Yang L, Shu Y, Zhang W, Zhang Y, Xu H. Single-cell transcriptomic analysis reveals gut microbiota-immunotherapy synergy through modulating tumor microenvironment. Signal Transduct Target Ther 2025; 10:140. [PMID: 40312419 PMCID: PMC12045981 DOI: 10.1038/s41392-025-02226-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 03/13/2025] [Accepted: 03/14/2025] [Indexed: 05/03/2025] Open
Abstract
The gut microbiota crucially regulates the efficacy of immune checkpoint inhibitor (ICI) based immunotherapy, but the underlying mechanisms remain unclear at the single-cell resolution. Using single-cell RNA sequencing and subsequent validations, we investigate gut microbiota-ICI synergy by profiling the tumor microenvironment (TME) and elucidating critical cellular interactions in mouse models. Our findings reveal that intact gut microbiota combined with ICIs may synergistically increase the proportions of CD8+, CD4+, and γδ T cells, reduce glycolysis metabolism, and reverse exhausted CD8+ T cells into memory/effector CD8+ T cells, enhancing antitumor response. This synergistic effect also induces macrophage reprogramming from M2 protumor Spp1+ tumor-associated macrophages (TAMs) to Cd74+ TAMs, which act as antigen-presenting cells (APCs). These macrophage subtypes show a negative correlation within tumors, particularly during fecal microbiota transplantation. Depleting Spp1+ TAMs in Spp1 conditional knockout mice boosts ICI efficacy and T cell infiltration, regardless of gut microbiota status, suggesting a potential upstream role of the gut microbiota and highlighting the crucial negative impact of Spp1+ TAMs during macrophage reprogramming on immunotherapy outcomes. Mechanistically, we propose a γδ T cell-APC-CD8+ T cell axis, where gut microbiota and ICIs enhance Cd40lg expression on γδ T cells, activating Cd40 overexpressing APCs (e.g., Cd74+ TAMs) through CD40-CD40L-related NF-κB signaling and boosting CD8+ T cell responses via CD86-CD28 interactions. These findings highlight the potential importance of γδ T cells and SPP1-related macrophage reprogramming in activating CD8+ T cells, as well as the synergistic effect of gut microbiota and ICIs in immunotherapy through modulating the TME.
Collapse
Affiliation(s)
- Minyuan Cao
- Department of Laboratory Medicine/Research Centre of Clinical Laboratory Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yun Deng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Qing Hao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Huayun Yan
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Quan-Lin Wang
- Department of Clinical Pharmacology, Xiangya Hospital, Central Laboratory of Hunan Cancer Hospital, Central South University, Changsha, China
| | - Chunyan Dong
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Jing Wu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yajiao He
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Li-Bin Huang
- Division of Gastrointestinal Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Xuyang Xia
- Department of Laboratory Medicine/Research Centre of Clinical Laboratory Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Division of Gastrointestinal Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Yongchao Gao
- Department of Clinical Pharmacology, Xiangya Hospital, Central Laboratory of Hunan Cancer Hospital, Central South University, Changsha, China
| | - Hai-Ning Chen
- Colorectal Cancer Center, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Wei-Han Zhang
- Gastric Cancer Center, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Yan-Jing Zhang
- Core Facilities, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaozhen Zhuo
- Department of Cardiology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Lunzhi Dai
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Hongbo Hu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yong Peng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Feng Zhang
- Center for Precision Medicine, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, China
| | - Zhaoqian Liu
- Department of Clinical Pharmacology, Xiangya Hospital, Central Laboratory of Hunan Cancer Hospital, Central South University, Changsha, China
| | - Weihua Huang
- Department of Clinical Pharmacology, Xiangya Hospital, Central Laboratory of Hunan Cancer Hospital, Central South University, Changsha, China
| | - Huiyuan Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Li Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yang Shu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Gastric Cancer Center, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Wei Zhang
- Department of Clinical Pharmacology, Xiangya Hospital, Central Laboratory of Hunan Cancer Hospital, Central South University, Changsha, China.
- The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China.
| | - Yan Zhang
- Lung Cancer Center/Lung Cancer Institute, Department of Medical Oncology, West China Hospital, Sichuan University, Chengdu, China.
| | - Heng Xu
- Department of Laboratory Medicine/Research Centre of Clinical Laboratory Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China.
- Institute of General Surgery, West China Hospital, Sichuan University, Chengdu, China.
- Tianfu Jincheng Laboratory, Chengdu, China.
| |
Collapse
|