1
|
Yang X, Li J, Xu C, Zhang G, Che X, Yang J. Potential mechanisms of rheumatoid arthritis therapy: Focus on macrophage polarization. Int Immunopharmacol 2024; 142:113058. [PMID: 39236455 DOI: 10.1016/j.intimp.2024.113058] [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: 07/18/2024] [Revised: 08/29/2024] [Accepted: 08/30/2024] [Indexed: 09/07/2024]
Abstract
Rheumatoid arthritis (RA) is an autoimmune inflammatory disease that affects multiple organs and systems in the human body, often leading to disability. Its pathogenesis is complex, and the long-term use of traditional anti-rheumatic drugs frequently results in severe toxic side effects. Therefore, the search for a safer and more effective antirheumatic drug is extremely important for the treatment of RA. As important immune cells in the body, macrophages are polarized. Under pathological conditions, macrophages undergo proliferation and are recruited to diseased tissues upon stimulation. In the local microenvironment, they polarize into different types of macrophages in response to specific factors and perform unique functions and roles. Previous studies have shown that there is a link between macrophage polarization and RA, indicating that certain active ingredients can ameliorate RA symptoms through macrophage polarization. Notably, Traditional Chinese medicine (TCM) monomer component and compounds demonstrate a particular advantage in this process. Building upon this insight, we reviewed and analyzed recent studies to offer valuable and meaningful insights and directions for the development and application of anti-rheumatic drugs.
Collapse
Affiliation(s)
- Xinyu Yang
- College of Acupuncture and Massage, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jinling Li
- College of Acupuncture and Massage, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Chengchao Xu
- College of Rehabilitation Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Guangheng Zhang
- Department of First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xinzhen Che
- Department of First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jiguo Yang
- College of Acupuncture and Massage, Shandong University of Traditional Chinese Medicine, Jinan, China.
| |
Collapse
|
2
|
Chang JW, Tang CH. The role of macrophage polarization in rheumatoid arthritis and osteoarthritis: Pathogenesis and therapeutic strategies. Int Immunopharmacol 2024; 142:113056. [PMID: 39217882 DOI: 10.1016/j.intimp.2024.113056] [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: 07/09/2024] [Revised: 08/17/2024] [Accepted: 08/30/2024] [Indexed: 09/04/2024]
Abstract
Rheumatoid arthritis (RA) and osteoarthritis (OA) are common and debilitating joint disorders affecting millions of individuals worldwide. Despite their distinct pathological features, both conditions share a crucial role of macrophages in disease progression. Macrophages exhibit remarkable plasticity, polarizing into pro-inflammatory M1 or anti-inflammatory M2 phenotypes in response to environmental cues. An imbalance in macrophage polarization, particularly a shift towards the M1 phenotype, contributes to chronic inflammation and joint damage in RA and OA. This review explores the complex interplay between macrophages and various cell types, including T cells, B cells, synovial fibroblasts, osteoclasts, chondrocytes, and adipocytes, in the pathogenesis of these diseases. We discuss the current understanding of macrophage polarization in RA and OA, highlighting the molecular mechanisms involved. Furthermore, we provide an overview of potential therapeutic strategies targeting macrophage polarization, such as disease-modifying anti-rheumatic drugs, traditional Chinese medicine, nanomedicines, proteins, chemical compounds, and physical therapies. By elucidating the precise mechanisms governing macrophage polarization and its interactions with other cells in the joint microenvironment, researchers can identify novel therapeutic targets and develop targeted interventions to alleviate disease progression and improve patient outcomes in RA and OA.
Collapse
Affiliation(s)
- Jun-Way Chang
- The Ph.D. Program of Biotechnology and Biomedical Industry, China Medical University, Taichung, Taiwan
| | - Chih-Hsin Tang
- Department of Pharmacology, School of Medicine, China Medical University, Taichung, Taiwan; Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan; Chinese Medicine Research Center, China Medical University, Taichung, Taiwan; Department of Medical Laboratory Science and Biotechnology, College of Medical and Health Science, Asia University, Taichung, Taiwan; Department of Medical Research, China Medical University Hsinchu Hospital, Hsinchu, Taiwan.
| |
Collapse
|
3
|
Pan X, Zong Q, Liu C, Wu H, Fu B, Wang Y, Sun W, Zhai Y. Konjac glucomannan exerts regulatory effects on macrophages and its applications in biomedical engineering. Carbohydr Polym 2024; 345:122571. [PMID: 39227106 DOI: 10.1016/j.carbpol.2024.122571] [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: 06/27/2024] [Revised: 07/22/2024] [Accepted: 07/31/2024] [Indexed: 09/05/2024]
Abstract
Konjac glucomannan (KGM) molecular chains contain a small amount of acetyl groups and a large number of hydroxyl groups, thereby exhibiting exceptional water retention and gel-forming properties. To meet diverse requirements, KGM undergoes modification processes such as oxidation, acetylation, grafting, and cationization, which reduce its viscosity, enhance its mechanical strength, and improve its water solubility. Researchers have found that KGM and its derivatives can regulate the polarization of macrophages, inducing their transformation into classically activated M1-type macrophages or alternatively activated M2-type macrophages, and even facilitating the interconversion between M1 and M2 phenotypes. Concurrently, the modulation of macrophage polarization states holds significant importance for chronic wound healing, inflammatory bowel disease (IBD), antitumor therapy, tissue engineering scaffolds, oral vaccines, pulmonary delivery, and probiotics. Therefore, KGM has the advantages of both immunomodulatory effects (biological activity) and gel-forming properties (physicochemical properties), giving it significant advantages in a variety of biomedical engineering applications.
Collapse
Affiliation(s)
- Xi Pan
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Qida Zong
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Chun Liu
- Hainan Institute for Drug Control, Haikou 570311, China
| | - Huiying Wu
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Bo Fu
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Ye Wang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Wei Sun
- Department of Biomedical Engineering, School of Pharmaceutical University, Shenyang 110016, China.
| | - Yinglei Zhai
- Department of Biomedical Engineering, School of Pharmaceutical University, Shenyang 110016, China.
| |
Collapse
|
4
|
Guan J, Wang J, Jia F, Jiang W, Song L, Xie L, Yang H, Han P, Lin H, Wu Z, Zhang X, Huang Y. Layer-by-layer self-assembly coatings on strontium titanate nanotubes with antimicrobial and anti-inflammatory properties to prevent implant-related infections. Colloids Surf B Biointerfaces 2024; 244:114183. [PMID: 39208607 DOI: 10.1016/j.colsurfb.2024.114183] [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: 06/15/2024] [Revised: 08/08/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
One way to effectively address endophyte infection and loosening is the creation of multifunctional coatings that combine anti-inflammatory, antibacterial, and vascularized osteogenesis. This study started with the preparation of strontium-doped titanium dioxide nanotubes (STN) on the titanium surface. Next, tannic acid (TA), gentamicin sulfate (GS), and pluronic F127 (PF127) were successfully loaded into the STN via layer-by-layer self-assembly, resulting in the STN@TA-GS/PF composite coatings. The findings demonstrated the excellent hydrophilicity and bioactivity of the STN@TA-GS/PF coating. STN@TA-GS/PF inhibited E. coli and S. aureus in vitro to a degree of roughly 80.95 % and 92.45 %, respectively. Cellular investigations revealed that on the STN@TA-GS/PF surface, the immune-system-related RAW264.7, the vasculogenic HUVEC, and the osteogenic MC3T3-E1 showed good adhesion and proliferation activities. STN@TA-GS/PF may influence RAW264.7 polarization toward the M2-type and encourage MC3T3-E1 differentiation toward osteogenesis at the molecular level. Meanwhile, the STN@TA-GS/PF coating achieved effective removal of ROS within HUVEC and significantly promoted angiogenesis. In both infected and non-infected bone defect models, the STN@TA-GS/PF material demonstrated strong anti-inflammatory, antibacterial, and vascularization-promoting osteogenesis properties. In addition, STN@TA-GS/PF had good hemocompatibility and biosafety. The three-step process used in this study to modify the titanium surface for several purposes gave rise to a novel concept for the clinical design of antimicrobial coatings with immunomodulatory properties.
Collapse
Affiliation(s)
- Jiaxin Guan
- College of Lab Medicine, Life Science Research Centre, Hebei North University, Zhangjiakou 075000, China
| | - Jiali Wang
- College of Lab Medicine, Life Science Research Centre, Hebei North University, Zhangjiakou 075000, China
| | - Fengzhen Jia
- College of Lab Medicine, Life Science Research Centre, Hebei North University, Zhangjiakou 075000, China
| | - Wenjing Jiang
- College of Lab Medicine, Life Science Research Centre, Hebei North University, Zhangjiakou 075000, China
| | - Lili Song
- College of Lab Medicine, Life Science Research Centre, Hebei North University, Zhangjiakou 075000, China
| | - Lei Xie
- School of Medicine, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Hao Yang
- Key Laboratory for Green Chemical Process of Ministry of Education, Wuhan Institute of Technology, Wuhan 430205, China
| | - Pengde Han
- School of Materials Science and Engineering, Yancheng Institute of Technology, Yancheng 224051, China
| | - He Lin
- School of Chemistry and Materials Science, Ludong University, Yantai 264025, China
| | - Zongze Wu
- Shenzhen Yakin Biotechnology Co., Shenzhen 518000, China.
| | - Xuejiao Zhang
- College of Lab Medicine, Life Science Research Centre, Hebei North University, Zhangjiakou 075000, China.
| | - Yong Huang
- College of Lab Medicine, Life Science Research Centre, Hebei North University, Zhangjiakou 075000, China.
| |
Collapse
|
5
|
Li X, Zhang G, Wang M, Lu C, Zhang G, Chen Z, Ji Y. Comparison of stromal vascular fraction cell composition between Coleman fat and extracellular matrix/stromal vascular fraction gel. Adipocyte 2024; 13:2360037. [PMID: 38829527 PMCID: PMC11152101 DOI: 10.1080/21623945.2024.2360037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 05/21/2024] [Indexed: 06/05/2024] Open
Abstract
As a mechanically condensed product of Coleman fat, extracellular matrix/stromal vascular fraction gel (ECM/SVF-gel) eliminates adipocytes, concentrates SVF cells, and improves fat graft retention. This study aims to compare SVF cell composition between Coleman fat and ECM/SVF-gel. Matched Coleman fat and ECM/SVF-gel of 28 healthy women were subjected to RNA-seq, followed by functional enrichment and cell-type-specific enrichment analyses, and deconvolution of SVF cell subsets, reconstructing SVF cell composition in the transcriptome level. ECM/SVF-gels had 9 upregulated and 73 downregulated differentially expressed genes (DEGs). Downregulated DEGs were mainly associated with inflammatory and immune responses, and enriched in fat macrophages. M2 macrophages, resting CD4+ memory T cells, M1 macrophages, resting mast cells, and M0 macrophages ranked in the top five most prevalent immune cells in the two groups. The proportions of the principal non-immune cells (e.g., adipose-derived stem cells, pericytes, preadipocytes, microvascular endothelial cells) had no statistical differences between the two groups. Our findings reveal ECM/SVF-gels share the same dominant immune cells beneficial to fat graft survival with Coleman fat, but exhibiting obvious losses of immune cells (especially macrophages), while non-immune cells necessary for adipose regeneration might have no significant loss in ECM/SVF-gels and their biological effects could be markedly enhanced by the ECM/SVF-gel's condensed nature.
Collapse
Affiliation(s)
- Xiaoyun Li
- Department of Pathology, Shantou University Medical College, Shantou, China
| | - Guohong Zhang
- Department of Pathology, Shantou University Medical College, Shantou, China
| | - Mengmeng Wang
- Medical Cosmetic Center, the First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Changhao Lu
- Department of Pathology, Shantou University Medical College, Shantou, China
| | - Guangping Zhang
- Department of Pathology, Shantou University Medical College, Shantou, China
| | - Zhehui Chen
- Medical Cosmetic Center, the First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Yingchang Ji
- Medical Cosmetic Center, the First Affiliated Hospital of Shantou University Medical College, Shantou, China
| |
Collapse
|
6
|
Zhou X, Guo YL, Xu C, Wang J. Macrophages: Key players in diabetic wound healing. World J Diabetes 2024; 15:2177-2181. [DOI: 10.4239/wjd.v15.i11.2177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 09/03/2024] [Accepted: 09/18/2024] [Indexed: 10/16/2024] Open
Abstract
In this editorial, we discuss the article by Wen et al published. Diabetic foot ulcers are prevalent and serious complications of diabetes, significantly impacting patients’ quality of life and often leading to disability or death, thereby placing a heavy burden on society. Effective diabetic wound healing is hindered by an imbalance in macrophage polarization; many macrophages fail to transition from the pro-inflammatory M1 phenotype to the anti-inflammatory M2 phenotype, which is crucial for tissue remodelling and repair. The wound healing process is both dynamic and complex. Healthy M1 macrophages, which have strong phagocytic abilities, are vital during the inflammatory phase of diabetic wound healing. However, the failure to transition to M2 macrophages during the proliferative phase hinders wound healing. We anticipate the development of new therapies that can repair damaged M1 macrophages during the inflammatory phase and promote M2 macrophage polarization during the proliferative phase, thereby enhancing the overall healing process.
Collapse
Affiliation(s)
- Xin Zhou
- Department of Science and Education, Huzhou Traditional Chinese Medicine Hospital Affiliated to Zhejiang Chinese Medical University, Huzhou 313000, Zhejiang Province, China
| | - Yan-Ling Guo
- Department of Ulcers and Peripheral Vascular Surgery, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture Moxibustion, Tianjin 300381, China
| | - Chuan Xu
- Department of Pharmacy, Huzhou Traditional Chinese Medicine Hospital Affiliated to Zhejiang Chinese Medical University, Huzhou 313000, Zhejiang Province, China
| | - Jun Wang
- Department of Ulcers and Peripheral Vascular Surgery, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture Moxibustion, Tianjin 300381, China
| |
Collapse
|
7
|
Purohit S, Mandal G, Biswas S, Dalui S, Gupta A, Chowdhury SR, Bhattacharyya A. AXL/GAS6 signaling governs differentiation of tumor-associated macrophages in breast cancer. Exp Cell Res 2024:114324. [PMID: 39510154 DOI: 10.1016/j.yexcr.2024.114324] [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: 04/28/2024] [Revised: 10/09/2024] [Accepted: 11/04/2024] [Indexed: 11/15/2024]
Abstract
Most epithelial cancers are infiltrated by prognostically relevant myelomonocytic cells. Immunosuppressive tumor associated macrophages (TAMs) and their precursor monocytic-MDSCs have previously been associated with worse outcomes in human breast cancer (BCa), yet the mechanism of immunosuppressive TAMs-polarization from myelomonocytic precursors is not completely understood. In this study, we show that persuaded AXL/GAS6 pathway alters macrophage phenotype from HLA-DRhighCD206low CD163low classical phagocytic into HLA-DRlowCD206highCD163high immunosuppressive ones with accelerated BCa progression, and increased angiogenesis signature and invasion ability of cancer cells at tumor beds. Notably, both AXL and GAS6 expressions are upregulated in human invasive breast carcinoma, with maximum expression in triple negative histology type. Mechanistically, we demonstrate that AXL/GAS6 signaling drives immunosuppression by governing increased immunosuppressive IL10 production while dampening IL-1β expression within the tumor microenvironment (TME) of BCa. Further, AXL/GAS6 signaling promotes angiogenesis through the activation of PI3K/AKT and NF-κB signaling pathways. Our results unveil role of AXL/GAS6 axis in the differentiation of TAMs, which governs malignant growth, and suggest that therapies that uncouple AXL/GAS6 axis may exhibit therapeutic opportunity for otherwise undruggable Triple Negative Breast Cancer (TNBC) patients.
Collapse
Affiliation(s)
- Suman Purohit
- Immunology Laboratory, Department of Zoology, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, West Bengal, India; Department of Zoology, Gurudas College, 1/1, Suren Sarkar Road, PhoolBagan, Kolkata 700054, West Bengal, India
| | - Gunjan Mandal
- Immunology Laboratory, Department of Zoology, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, West Bengal, India; Division of Cancer Biology, DBT-Institute of Life Sciences, Bhubaneswar 751023, India
| | - Subir Biswas
- Immunology Laboratory, Department of Zoology, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, West Bengal, India; Tumor Immunology and Immunotherapy, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai 410210, India; Homi Bhabha National Institute, Anushaktinagar, Mumbai 400 094, Maharashtra, India
| | - Shauryabrota Dalui
- Immunology Laboratory, Department of Zoology, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, West Bengal, India
| | - Arnab Gupta
- Department of Surgical Oncology, Saroj Gupta Cancer Centre and Research Institute, Mahatma Gandhi Road, Kolkata 700063, West Bengal, India
| | - Sougata Roy Chowdhury
- Immunology Laboratory, Department of Zoology, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, West Bengal, India; Translational Immunology Laboratory, Department of Life Science and Biotechnology, Jadavpur University, Kolkata 700032, West Bengal, India
| | - Arindam Bhattacharyya
- Immunology Laboratory, Department of Zoology, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, West Bengal, India.
| |
Collapse
|
8
|
Wang PH, Wang Y, Guo YY, Ma ZH, Wu C, Xing L. Ibuprofen modulates macrophage polarization by downregulating poly (ADP-ribose) polymerase 1. Int Immunopharmacol 2024; 143:113502. [PMID: 39488918 DOI: 10.1016/j.intimp.2024.113502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 09/21/2024] [Accepted: 10/22/2024] [Indexed: 11/05/2024]
Abstract
Ibuprofen, a non-steroidal drug, is well known for its anti-inflammatory activity. The effects of ibuprofen on the polarization of macrophages are still not clear. Herein, we used THP-1 monocyte-derived macrophages to find that ibuprofen has inhibitory effects on the polarization of both classically activated M1 macrophages and alternatively activated M2 macrophages by downregulating NF-κB and JAK/STAT signaling pathways. During M1 or M2 polarization, ibuprofen also downregulated the expression of poly (ADP-ribose) polymerase 1 (PARP1). Furthermore, knockdown of PARP1 by either small interfering RNA or PARP1 inhibitor PJ34 can exert inhibitory effects on the polarization of M1 and M2, and alter the immune response of macrophages to the infection of Mycobacterium tuberculosis H37Ra. The results demonstrate that PARP1 plays a regulatory role in the ibuprofen-modulated polarization of macrophage, revealing the interplay between the DNA repair response process and macrophage polarization.
Collapse
Affiliation(s)
- Pei-Hua Wang
- Institutes of Biomedical Sciences, Shanxi University, 92 Wucheng Road, Taiyuan 030006, Shanxi Province, China
| | - Yan Wang
- Institutes of Biomedical Sciences, Shanxi University, 92 Wucheng Road, Taiyuan 030006, Shanxi Province, China
| | - Yan-Yan Guo
- Institutes of Biomedical Sciences, Shanxi University, 92 Wucheng Road, Taiyuan 030006, Shanxi Province, China
| | - Zi-Hui Ma
- Institutes of Biomedical Sciences, Shanxi University, 92 Wucheng Road, Taiyuan 030006, Shanxi Province, China
| | - Changxin Wu
- Institutes of Biomedical Sciences, Shanxi University, 92 Wucheng Road, Taiyuan 030006, Shanxi Province, China; Shanxi Provincial Key Laboratory of Medical Molecular Cell Biology, Shanxi University, 92 Wucheng Road, Taiyuan 030006, China
| | - Li Xing
- Institutes of Biomedical Sciences, Shanxi University, 92 Wucheng Road, Taiyuan 030006, Shanxi Province, China; Shanxi Provincial Key Laboratory of Medical Molecular Cell Biology, Shanxi University, 92 Wucheng Road, Taiyuan 030006, China.
| |
Collapse
|
9
|
Li S, Fu W, Wang L. Role of macrophages in aortic dissection pathogenesis: insights from preclinical studies to translational prospective. SCIENCE CHINA. LIFE SCIENCES 2024; 67:2354-2367. [PMID: 39358669 DOI: 10.1007/s11427-024-2693-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 07/25/2024] [Indexed: 10/04/2024]
Abstract
Aortic dissection is a critical vascular disease that is characterized by a high mortality rate and inflammation significantly influences its onset and progression. Recent studies highlight the integral role of macrophages, key players in the immune system, in the pathological landscape of aortic dissection. These cells are involved in crucial processes, such as the remodeling of the extracellular matrix, immunocyte infiltration, and phenotypic switching of smooth muscle cells, which are essential for the structural integrity and functional dynamics of the aortic wall. Despite these insights, the specific contributions of macrophages to the development and progression of aortic dissection remains unclear. This review explores the pathogenesis of aortic dissection with a focus on macrophages and describes their origins, phenotypic variations, and potential roles based on the most recent research findings. Furthermore, we discuss key molecules related to macrophages during aortic dissection, their interactions with other cellular components within the aorta, and the implications of these interactions for future therapeutic strategies. This comprehensive analysis aimed to improve our understanding of macrophages in aortic dissection and promote the development of targeted interventions.
Collapse
Affiliation(s)
- Shiyi Li
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Vascular Surgery Institute of Fudan University, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
| | - Weiguo Fu
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Vascular Surgery Institute of Fudan University, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
- Key Laboratory of Panvascular Disease Precision Medicine, Zhongshan Hospital Xiamen, Fudan University, Xiamen, 361015, China
| | - Lixin Wang
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
- Vascular Surgery Institute of Fudan University, Shanghai, 200032, China.
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China.
- Key Laboratory of Panvascular Disease Precision Medicine, Zhongshan Hospital Xiamen, Fudan University, Xiamen, 361015, China.
- Fudan Zhangjiang Institute, Shanghai, 201203, China.
| |
Collapse
|
10
|
Wen JY, Fang YY, Li DM, Tang YL, Huang HQ, Liu LM, Zeng JH, Dang YW, Pan YF, Zeng DT, Huang WJ, Chen G, Li H. A Comprehensive Analysis of LYAR in Colorectal Cancer: Prognostic Marker and Therapeutic Target. Cancer Biother Radiopharm 2024; 39:673-689. [PMID: 39159060 DOI: 10.1089/cbr.2023.0181] [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: 08/21/2024] Open
Abstract
Background: Colorectal cancer (CRC) is a major global health challenge with a need for new biomarkers and therapeutic targets. This work investigated the biological mechanisms and clinical value of Ly1 antibody reactive (LYAR) in CRC. Methods: We analyzed LYAR mRNA expression across multiple public databases, including genotype-tissue expression, gene expression omnibus, Oncomine, and the cancer genome atlas, alongside in-house immunohistochemical data to evaluate LYAR protein expression in CRC and non-CRC colorectal tissues. Gene set enrichment analysis (GSEA) was used to elucidate LYAR's biological functions, and its impact on the tumor immune microenvironment was assessed using CIBERSORT, ESTIMATE, and single-cell RNA sequencing techniques. In addition, LYAR's association with clinicopathological features and patient prognosis was explored, and its influence on drug sensitivity was investigated using the Connectivity Map database. Results: LYAR was significantly upregulated in CRC tissues compared with non-CRC colorectal counterparts, associated with altered immune cell composition and enhanced RNA processing, splicing, and cell cycle regulation. High LYAR expression correlated with poor disease-free and overall survival, underscoring its prognostic value. GSEA revealed LYAR's involvement in critical cellular processes and pathways, including DNA repair, cell cycle, and mTORC1 signaling. Correlation analysis identified genes positively and negatively associated with LYAR, leading to the discovery of temsirolimus and WYE-354, mTOR inhibitors, as potential therapeutic agents for CRC. Furthermore, LYAR expression predicted increased sensitivity to cetuximab in RAS wild-type metastatic CRC, indicating its utility as a biomarker for treatment responsiveness. Conclusions: LYAR's upregulation in CRC highlights its potential as a biomarker for prognosis and therapeutic targeting, offering insights into CRC pathology and suggesting new avenues for treatment optimization.
Collapse
Affiliation(s)
- Jia-Ying Wen
- Department of Radiotherapy, The Second Affiliated Hospital of Guangxi Medical University, Nanning, P. R. China
| | - Ye-Ying Fang
- Department of Radiotherapy, The First Affiliated Hospital of Guangxi Medical University, Nanning, P. R. China
| | - Dong-Ming Li
- Department of Pathology, Guangxi Key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, The First Affiliated Hospital of Guangxi Medical University, Nanning, P. R. China
| | - Yu-Lu Tang
- Department of Pathology, Guangxi Key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, The First Affiliated Hospital of Guangxi Medical University, Nanning, P. R. China
| | - He-Qing Huang
- Department of Radiotherapy, The First Affiliated Hospital of Guangxi Medical University, Nanning, P. R. China
| | - Li-Min Liu
- Department of Toxicology, College of Pharmacy, Guangxi Medical University, Nanning, P.R. China
| | - Jiang-Hui Zeng
- Department of Clinical Laboratory, The Third Affiliated Hospital of Guangxi Medical University/Nanning Second People's Hospital, Nanning, P. R. China
| | - Yi-Wu Dang
- Department of Pathology, Guangxi Key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, The First Affiliated Hospital of Guangxi Medical University, Nanning, P. R. China
| | - Yan-Fang Pan
- Department of Pathology, Hospital of Guangxi Liugang Medical Co., LTD./Guangxi Liuzhou Dingshun Forensic Expert Institute, Liuzhou, P.R. China
| | - Da-Tong Zeng
- Department of Pathology, Redcross Hospital of Yulin city, Yulin, P.R. China
| | - Wei-Jian Huang
- Department of Pathology, Redcross Hospital of Yulin city, Yulin, P.R. China
| | - Gang Chen
- Department of Pathology, Guangxi Key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, The First Affiliated Hospital of Guangxi Medical University, Nanning, P. R. China
| | - Hui Li
- Department of Colorectal & Anal Surgery, Guangxi Key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, The First Affiliated Hospital of Guangxi Medical University, Nanning, P. R. China
| |
Collapse
|
11
|
Yang YF, Cheng SY, Wang YL, Yue ZP, Yu YX, Chen YZ, Wang WK, Xu ZR, Qi ZQ, Liu Y. Accumulated inflammation and fibrosis participate in atrazine induced ovary toxicity in mice. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 360:124672. [PMID: 39103034 DOI: 10.1016/j.envpol.2024.124672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 07/24/2024] [Accepted: 08/03/2024] [Indexed: 08/07/2024]
Abstract
Atrazine is a widely used herbicide in agricultural production. Previous studies have shown that atrazine affects hormone secretion and oocyte maturation in female reproduction. However, the specific mechanism by which atrazine affects ovarian function remains unclear. In this study, using a mouse gastric lavage model, we report that four weeks of atrazine exposure affects body growth, interferes with the estrous cycle, and increases the number of atretic follicles in mice. The expression levels of follicle development related factors StAR, BMP15, and AMH decreased. Metabolomic analysis revealed that atrazine activates an inflammatory response in ovarian tissue. Further studies confirmed that the expression levels of TNF-α, IL-6, and NF-κB increased in the ovaries of mice exposed to atrazine. Additionally, α-smooth muscle actin (α-SMA) accumulated in ovarian tissue, and transforming growth factor-β (TGF-β) signaling was activated, indicating the occurrence of tissue fibrosis. Moreover, mice exposed to atrazine produced fewer oocytes and exhibited reduced embryonic development. Furthermore, mice exposed to atrazine exhibited altered gut microbiota abundance and a disrupted colon barrier. Collectively, these findings suggest that atrazine exposure induces ovarian inflammation and fibrosis, disrupts ovarian homeostasis, and impairs follicle maturation, ultimately reducing oocyte quality.
Collapse
Affiliation(s)
- Yi-Fan Yang
- Medical College, Guangxi University, Nanning, Guangxi, 530004, China
| | - Si-Yao Cheng
- Medical College, Guangxi University, Nanning, Guangxi, 530004, China
| | - Ya-Long Wang
- Center for Reproductive Medicine, Maternity and Child Health Care Hospital in Xiangtan, Xiangtan, Hunan, 411100, China
| | - Zhao-Ping Yue
- Center for Reproductive Medicine, Maternity and Child Health Care Hospital in Xiangtan, Xiangtan, Hunan, 411100, China
| | - Yu-Xi Yu
- Medical College, Guangxi University, Nanning, Guangxi, 530004, China
| | - Yan-Zhu Chen
- Medical College, Guangxi University, Nanning, Guangxi, 530004, China
| | - Wen-Ke Wang
- Medical College, Guangxi University, Nanning, Guangxi, 530004, China
| | - Zhi-Ran Xu
- Translational Medicine Research Center, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, Guangxi, 530011, China
| | - Zhong-Quan Qi
- Medical College, Guangxi University, Nanning, Guangxi, 530004, China
| | - Yu Liu
- Medical College, Guangxi University, Nanning, Guangxi, 530004, China.
| |
Collapse
|
12
|
Shen C, Deng M, Wang X, Li X, Chen X, Gao Z, Li C, Liu Y. Zuogui Wan modulates macrophage polarization and promotes osteogenic differentiation through regulation of CD51-positive bone marrow mesenchymal stem cells. Sci Rep 2024; 14:26130. [PMID: 39478130 PMCID: PMC11525575 DOI: 10.1038/s41598-024-77590-5] [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: 03/28/2024] [Accepted: 10/23/2024] [Indexed: 11/02/2024] Open
Abstract
Background Zuogui Wan (ZGW) is a traditional herbal formula used to treat chronic kidney and bone diseases. Previous research has shown that ZGW slows down the aging process of bone marrow mesenchymal stem cells (BMSCs) and improves bone metabolism. However, its role in treating postmenopausal osteoporosis (PMOP) has not yet been fully investigated. Therefore, we investigated the therapeutic effects of ZGW and its potential mechanisms in an ovariectomy (OVX)-induced osteoporosis rat model. Results We observed significant improvements in bone loss and the osteoporotic phenotype in OVX rats treated with ZGW. These findings were confirmed with micro-computed tomography (micro-CT) and histomorphological analysis. We also discovered that ZGW reversed the macrophage imbalance, which in turn inhibited osteoclast differentiation and bone resorption. Furthermore, RNA-Seq results revealed the active expression of CD51 in BMSCs before and after ZGW therapy, which is associated with macrophage polarization and osteoblastic differentiation. The results also showed that ZGW decreased CD51 + BMSCs levels, which is closely related to the inhibition of osteoblast differentiation and promotion of osteoclast resorption. Conclusions Our study demonstrated that ZGW may improve postmenopausal osteoporosis by restoring macrophage polarization and down-regulating CD51 + BMSCs. In addition, ZGW promoted osteoblast formation and inhibited osteoclast resorption.
Collapse
Affiliation(s)
- Chongyang Shen
- Basic Medicine School, Chengdu University of Traditional Chinese Medicine, Chengdu, People's Republic of China
| | - Mingxing Deng
- Basic Medicine School, Chengdu University of Traditional Chinese Medicine, Chengdu, People's Republic of China
| | - Xiaobao Wang
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, People's Republic of China
| | - Xiaoyu Li
- Basic Medicine School, Chengdu University of Traditional Chinese Medicine, Chengdu, People's Republic of China
| | - Xiongbin Chen
- Basic Medicine School, Chengdu University of Traditional Chinese Medicine, Chengdu, People's Republic of China
| | - Zhi Gao
- Sichuan Orthopedic Hospital, Chengdu, People's Republic of China
| | - Chuncai Li
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, People's Republic of China.
| | - Yincong Liu
- Basic Medicine School, Chengdu University of Traditional Chinese Medicine, Chengdu, People's Republic of China.
| |
Collapse
|
13
|
Voskoboynik Y, McCulloch AD, Sahoo D. Macrophages on the run: Exercise balances macrophage polarization for improved health. Mol Metab 2024; 90:102058. [PMID: 39476967 DOI: 10.1016/j.molmet.2024.102058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 10/11/2024] [Accepted: 10/24/2024] [Indexed: 11/09/2024] Open
Abstract
OBJECTIVE Exercise plays a crucial role in maintaining and improving human health. However, the precise molecular mechanisms that govern the body's response to exercise or/compared to periods of inactivity remain elusive. Current evidence appears to suggest that exercise exerts a seemingly dual influence on macrophage polarization states, inducing both pro-immune response M1 activation and cell-repair-focused M2 activation. To reconcile this apparent paradox, we leveraged a comprehensive meta-analysis of 75 diverse exercise and immobilization published datasets (7000+ samples), encompassing various exercise modalities, sampling techniques, and species. METHODS 75 exercise and immobilization expression datasets were identified and processed for analysis. The data was analyzed using boolean relationships which uses binary gene expression relationships in order to increase the signal to noise achieved from the data, allowing for the use of comparison across such a diverse set of datasets. We utilized a boolean relationship-aided macrophage gene model [1], to model the macrophage polarization state in pre and post exercise samples in both immediate exercise and long term training. RESULTS Our modeling uncovered a key temporal dynamic: exercise triggers an immediate M1 surge, while long term training transitions to sustained M2 activation. These patterns were consistent across different species (human vs mouse), sampling methods (blood vs muscle biopsy), and exercise type (resistance vs endurance), and routinely showed statistically significant results. Immobilization was shown to have the opposite effect of exercise by triggering an immediate M2 activation. Individual characteristics like gender, exercise intensity and age were found to impact the degree of polarization without changing the overall patterns. To model macrophages within the specific context of muscle tissue, we identified a focused gene set signature of muscle resident macrophage polarization, allowing for the precise measurement of macrophage activity in response to exercise within the muscle. CONCLUSIONS These consistent patterns across all 75 examined studies suggest that the long term health benefits of exercise stem from its ability to orchestrate a balanced and temporally-regulated interplay between pro-immune response (M1) and reparative macrophage activity (M2). Similarly, it suggests that an imbalance between pro-immune and cell repair responses could facilitate disease development. Our findings shed light on the intricate molecular choreography behind exercise-induced health benefits with a particular insight on its effect on the macrophages within the muscle.
Collapse
Affiliation(s)
- Yotam Voskoboynik
- Department of Bioinformatics and System Biology, Jacobs School of Engineering, University of California San Diego, San Diego, United States
| | - Andrew D McCulloch
- Department of Bioengineering, University of California San Diego, United States; Department of Medicine, University of California San Diego, United States
| | - Debashis Sahoo
- Department of Pediatrics, University of California San Diego, United States; Department of Computer Science and Engineering, Jacob's School of Engineering, University of California San Diego, United States.
| |
Collapse
|
14
|
Wang J, Wang L, Han L, Han Y, Gu J, Chen Z. Formononetin attenuates hepatic injury in diabetic mice by regulating macrophage polarization through the PTP1B/STAT6 axis. Int Immunopharmacol 2024; 140:112802. [PMID: 39088924 DOI: 10.1016/j.intimp.2024.112802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 07/05/2024] [Accepted: 07/23/2024] [Indexed: 08/03/2024]
Abstract
BACKGROUND Formononetin (FNT) is an isoflavone known for its anti-inflammatory properties and has been shown to reduce insulin resistance in Type 2 Diabetes Mellitus (T2DM). However, its effects and the underlying mechanisms in diabetic liver injury remain largely unexplored. METHODS We established a T2DM-induced liver injury mouse model by feeding high-fat diet, followed by injecting streptozotocin. The mice were then treated with FNT and the liver function in these mice was assessed. Macrophage markers in FNT-treated T2DM mice or human THP-1 cells were evaluated using flow cytometry, RT-qPCR, and Western blotting. The expression of PTP1B and STAT6 in mouse liver tissues and THP-1 cells was analyzed. Molecular docking predicted the interaction between PTP1B and STAT6, which was validated via co-immunoprecipitation (Co-IP) and phos-tag analysis. Microscale thermophoresis (MST) assessed the binding affinity of FNT to PTP1B. RESULTS FNT treatment significantly ameliorated blood glucose levels, hepatocyte apoptosis, inflammatory response, and liver dysfunction in T2DM mice. Moreover, FNT facilitated M2 macrophage polarization in both T2DM mice and high glucose (HG)-induced THP-1-derived macrophages. The PTP1B/STAT6 axis, deregulated in T2DM mice, was normalized by FNT treatment, which counteracted the T2DM-induced upregulation of PTP1B and downregulation of phosphorylated STAT6. Molecular docking and subsequent analyses revealed that PTP1B binds to and dephosphorylates STAT6 at the S325A site. In contrast, FNT strongly binds to PTP1B and influences its expression at the K116A site, promoting M2 polarization of THP-1 cells via downregulation of PTP1B. CONCLUSION Formononetin mitigates diabetic hepatic injury by fostering M2 macrophage polarization via the PTP1B/STAT6 axis.
Collapse
Affiliation(s)
- Jinchun Wang
- Department of Pharmacy, Jiangsu Health Vocational College, 150 Fenghuang W St, Gulou, Nanjing, Jiangsu 211800, China
| | - Lei Wang
- Department of Pharmacy, Jiangsu Health Vocational College, 150 Fenghuang W St, Gulou, Nanjing, Jiangsu 211800, China
| | - Lei Han
- Department of Pharmacy, Jiangsu Health Vocational College, 150 Fenghuang W St, Gulou, Nanjing, Jiangsu 211800, China
| | - Yiwen Han
- Department of Clinical Medicine, Jiangsu Health Vocational College, 150 Fenghuang W St, Gulou, Nanjing, Jiangsu 211800, China
| | - Jun Gu
- Department of Public Health, Nanjing Medical University, 140 Hanzhong Rd, Gulou, Nanjing, Jiangsu 211166, China
| | - Zhujing Chen
- Department of Outpatient, Jurong People's Hospital, Jurong, No 66. Two holy road, Jurong, Zhenjiang, Jiangsu 212400, China.
| |
Collapse
|
15
|
Kushwaha N, Panjwani D, Patel S, Ahlawat P, Yadav MR, Patel AS. Emerging advances in nano-biomaterial assisted amyloid beta chimeric antigen receptor macrophages (CAR-M) therapy: reducing plaque burden in Alzheimer's disease. J Drug Target 2024:1-21. [PMID: 39403775 DOI: 10.1080/1061186x.2024.2417012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 09/23/2024] [Accepted: 10/08/2024] [Indexed: 10/22/2024]
Abstract
Alzheimer's disease is the most common form, accounting for 60-70% of 55 million dementia cases. Even though the precise pathophysiology of AD is not completely understood, clinical trials focused on antibodies targeting aggregated forms of β amyloid (Aβ) have demonstrated that reducing amyloid plaques can arrest cognitive decline in patients in the early stages of AD. In this study, we provide an overview of current research and innovations for controlled release from nano-biomaterial-assisted chimeric antigen receptor macrophage (CAR-M) therapeutic strategies targeted at AD. Nano-bio materials, such as iron-oxide nanoparticles (IONPs), can be made selectively (Hp-Hb/mannose) to bind and take up Aβ plaques like CAR-M cells. By using nano-bio materials, both the delivery and stability of CAR-M cells in brain tissue can be improved to overcome the barriers of the BBB and enhance therapeutic effects. By enhancing the targeting capabilities and stability of CAR-M cells, mRNA-loaded nano-biomaterials can significantly improve the efficacy of immunotherapy for plaque reduction in AD. This novel strategy holds promise for translating preclinical successes into clinical applications, potentially revolutionising the management of AD.
Collapse
Affiliation(s)
- Nishabh Kushwaha
- Department of Pharmaceutics, Parul Institute of Pharmacy, Parul University, Vadodara, India
| | - Drishti Panjwani
- Department of Pharmaceutics, Parul Institute of Pharmacy, Parul University, Vadodara, India
| | - Shruti Patel
- Department of Pharmaceutics, Parul Institute of Pharmacy, Parul University, Vadodara, India
| | - Priyanka Ahlawat
- Department of Pharmaceutics, Parul Institute of Pharmacy, Parul University, Vadodara, India
| | - Mange Ram Yadav
- Research and Development Cell, Parul University, Vadodara, India
| | - Asha S Patel
- Department of Pharmaceutics, Parul Institute of Pharmacy, Parul University, Vadodara, India
| |
Collapse
|
16
|
Luo Y, Hu S, Li Y, Ma L. Inflammation environment-adaptive matrix confinement for three-dimensional modulation of macrophages. Biomater Sci 2024; 12:5324-5336. [PMID: 39248106 DOI: 10.1039/d4bm00939h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/10/2024]
Abstract
The balance of macrophages in immune reactions is crucial for tissue repair. Despite some studies on responsive surfaces for immunomodulation regulation of macrophage phenotypes via external stimuli, 2D and manual interventions are limited. Herein, to address these limitations, we developed an inflammation environment-responsive macrophage-laden hydrogel-filled scaffold for investigating the impact of matrix confinement on macrophage phenotypes adaptively. We fabricated gelatin scaffolds with a controllable pore size and found that macrophages within smaller pores tended to have an anti-inflammation phenotype. We prepared poly(vinyl alcohol) (PVA)-based hydrogels crosslinked with phenylboronic acid (PBA)-based linkers. The hydrogels possessed shear-thinning, cell-loading, and ROS-sensitive-degradation abilities. Subsequently, a macrophage-laden hydrogel-filled scaffold was fabricated by filling the hydrogels into the porous scaffold under vacuum. With the degradation of the hydrogels under the overexpression of ROS in an inflammation environment, the macrophages were transferred from a state with strong matrix confinement to that with a weaker one. Meanwhile, with the change in matrix confinement, the macrophages upregulated the expressions of Arg-1 and IL-10 and downregulated the expressions of IL-1β, TNF-α, and IL-6, indicating polarization toward the anti-inflammatory phenotype. The inflammation environment-adaptive modulation of macrophage phenotypes in 3D provides a smart and biomimetic strategy for immunomodulation and regenerative medicine.
Collapse
Affiliation(s)
- Yilun Luo
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China.
- MOE Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, School of Medical Information Engineering, Gannan Medical University, Ganzhou 341000, China
| | - Sentao Hu
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China.
| | - Yan Li
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China.
| | - Lie Ma
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China.
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China
| |
Collapse
|
17
|
Huang T, Bei C, Hu Z, Li Y. CAR-macrophage: Breaking new ground in cellular immunotherapy. Front Cell Dev Biol 2024; 12:1464218. [PMID: 39421021 PMCID: PMC11484238 DOI: 10.3389/fcell.2024.1464218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 09/23/2024] [Indexed: 10/19/2024] Open
Abstract
Chimeric Antigen Receptor (CAR) technology has revolutionized cellular immunotherapy, particularly with the success of CAR-T cells in treating hematologic malignancies. However, CAR-T cells have the limited efficacy of against solid tumors. To address these limitations, CAR-macrophages (CAR-Ms) leverage the innate properties of macrophages with the specificity and potency of CAR technology, offering a novel and promising approach to cancer immunotherapy. Preclinical studies have shown that CAR-Ms can effectively target and destroy tumor cells, even within challenging microenvironments, by exhibiting direct cytotoxicity and enhancing the recruitment and activation of other immune cells. Additionally, the favorable safety profile of macrophages and their persistence within solid tumors position CAR-Ms as potentially safer and more durable therapeutic options compared to CAR-T cells. This review explores recent advancements in CAR-Ms technology, including engineering strategies to optimize their anti-tumor efficacy and preclinical evidence supporting their use. We also discuss the challenges and future directions in developing CAR-Ms therapies, emphasizing their potential to revolutionize cellular immunotherapy. By harnessing the unique properties of macrophages, CAR-Ms offer a groundbreaking approach to overcoming the current limitations of CAR-T cell therapies, paving the way for more effective and sustainable cancer treatments.
Collapse
Affiliation(s)
- Ting Huang
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Chenqi Bei
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhenhua Hu
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, China
| | - Yuanyuan Li
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
18
|
Galué-Parra A, de Moraes LS, Hage AAP, Castro de Sena CB, Nascimento JLMD, da Silva EO. In vitro immunomodulatory effects of Caryocar villosum oil on murine macrophages. Biomed Pharmacother 2024; 179:117360. [PMID: 39232387 DOI: 10.1016/j.biopha.2024.117360] [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: 07/04/2024] [Revised: 08/16/2024] [Accepted: 08/26/2024] [Indexed: 09/06/2024] Open
Abstract
Macrophages undergo activation in response to multiple stimuli, including pathogens, growth factors and natural products. The inflammatory response and oxidative stress play critical roles in such macrophage activation. Some natural products reportedly promote immunoregulatory effects and the control of macrophage activation. Caryocar villosum (Cv), a native amazon plant, contains compounds that are an important source of molecules capable of macrophage activation. Herein, we demonstrate the immunomodulatory effects of oil obtained from Caryocar villosum (CvO) on macrophages. Macrophages were treated with varying concentrations of CvO, and resulting cellular morphological and functional changes were evaluated, including the production of nitric oxide (NO), reactive oxygen species (ROS), cytokines and phagocytic activity. Treatment of cells with 50 and 100 μg/mL CvO induced morphological and physiological alterations in the macrophages, such as increased cell surface and phagocytic activity. Additionally, treatment increased the productions of inflammatory cytokines (INF-γ, TNF-α, IL-6) and anti-inflammatory cytokines (IL-17 and IL-10) by macrophages, and significantly decreased ROS levels. In conclusion, these data suggest that, due to molecular diversity, CvO promoted an immunomodulatory effect on macrophages, mediated by an increased production of cytokines, and inhibition of ROS generation and phagocytic activity. Thus, CvO presents potential as a therapeutic agent for the treatment of inflammatory and non-inflammatory diseases.
Collapse
Affiliation(s)
- Adan Galué-Parra
- Laboratory of Structural Biology, Federal University of Para Institute of Biological Sciences, Belém, Pará, Brazil; Postgraduate Program in Biology of Infectious and Parasitic Agents, Federal University of Para Institute of Biological Sciences, Belém, Pará, Brazil; National Institute of Science and Technology in Structural Biology and Bioimaging, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Lienne Silveira de Moraes
- Laboratory of Structural Biology, Federal University of Para Institute of Biological Sciences, Belém, Pará, Brazil
| | - Amanda Anastácia Pinto Hage
- Laboratory of Structural Biology, Federal University of Para Institute of Biological Sciences, Belém, Pará, Brazil
| | - Chubert Bernardo Castro de Sena
- Laboratory of Structural Biology, Federal University of Para Institute of Biological Sciences, Belém, Pará, Brazil; National Institute of Science and Technology in Neuroimmunomodulation (INCT-NIM), Rio de Janeiro, Rio de Janeiro, Brazil
| | - Jose Luiz Martins do Nascimento
- Laboratory of Molecular and Cellular Neurochemistry, Federal University of Para Institute of Biological Sciences, Belém, Pará, Brazil; National Institute of Science and Technology in Neuroimmunomodulation (INCT-NIM), Rio de Janeiro, Rio de Janeiro, Brazil
| | - Edilene Oliveira da Silva
- Laboratory of Structural Biology, Federal University of Para Institute of Biological Sciences, Belém, Pará, Brazil; Postgraduate Program in Biology of Infectious and Parasitic Agents, Federal University of Para Institute of Biological Sciences, Belém, Pará, Brazil; National Institute of Science and Technology in Structural Biology and Bioimaging, Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
19
|
Yang L, Fu MF, Wang HY, Sun H. Research Advancements in the Interplay between T3 and Macrophages. Curr Med Sci 2024; 44:883-889. [PMID: 39446284 DOI: 10.1007/s11596-024-2935-6] [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: 12/14/2023] [Accepted: 09/03/2024] [Indexed: 10/25/2024]
Abstract
3,3',5-Triiodo-L-thyronine (T3) is a key endocrine hormone in the human body that plays crucial roles in growth, development, metabolism, and immune function. Macrophages, the key regulatory cells within the immune system, exhibit marked "heterogeneity" and "plasticity", with their phenotype and function subject to modulation by local environmental signals. The interplay between the endocrine and immune systems is well documented. Numerous studies have shown that T3 significantly target macrophages, highlighting them as key cellular components in this interaction. Through the regulation of macrophage function and phenotype, T3 influences immune function and tissue repair in the body. This review comprehensively summarizes the regulatory actions and mechanisms of T3 on macrophages, offering valuable insights into further research of the immunoregulatory effects of T3.
Collapse
Affiliation(s)
- Liu Yang
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Provincial Clinical Research Center for Diabetes and Metabolic Disorders, Wuhan, 430022, China
| | - Meng-Fei Fu
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Provincial Clinical Research Center for Diabetes and Metabolic Disorders, Wuhan, 430022, China
| | - Han-Yu Wang
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Provincial Clinical Research Center for Diabetes and Metabolic Disorders, Wuhan, 430022, China
| | - Hui Sun
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Provincial Clinical Research Center for Diabetes and Metabolic Disorders, Wuhan, 430022, China.
| |
Collapse
|
20
|
Kang M, Jia H, Feng M, Ren H, Gao J, Liu Y, Zhang L, Zhou MS. Cardiac macrophages in maintaining heart homeostasis and regulating ventricular remodeling of heart diseases. Front Immunol 2024; 15:1467089. [PMID: 39372400 PMCID: PMC11449765 DOI: 10.3389/fimmu.2024.1467089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 09/03/2024] [Indexed: 10/08/2024] Open
Abstract
Macrophages are most important immune cell population in the heart. Cardiac macrophages have broad-spectrum and heterogeneity, with two extreme polarization phenotypes: M1 pro-inflammatory macrophages (CCR2-ly6Chi) and M2 anti-inflammatory macrophages (CCR2-ly6Clo). Cardiac macrophages can reshape their polarization states or phenotypes to adapt to their surrounding microenvironment by altering metabolic reprogramming. The phenotypes and polarization states of cardiac macrophages can be defined by specific signature markers on the cell surface, including tumor necrosis factor α, interleukin (IL)-1β, inducible nitric oxide synthase (iNOS), C-C chemokine receptor type (CCR)2, IL-4 and arginase (Arg)1, among them, CCR2+/- is one of most important markers which is used to distinguish between resident and non-resident cardiac macrophage as well as macrophage polarization states. Dedicated balance between M1 and M2 cardiac macrophages are crucial for maintaining heart development and cardiac functional and electric homeostasis, and imbalance between macrophage phenotypes may result in heart ventricular remodeling and various heart diseases. The therapy aiming at specific target on macrophage phenotype is a promising strategy for treatment of heart diseases. In this article, we comprehensively review cardiac macrophage phenotype, metabolic reprogramming, and their role in maintaining heart health and mediating ventricular remodeling and potential therapeutic strategy in heart diseases.
Collapse
Affiliation(s)
- Mengjie Kang
- Science and Experiment Research Center, Shenyang Medical College & Shenyang Key Laboratory of Vascular Biology, Science and Experimental Research Center, Shenyang Medical College, Shenyang, China
| | - Hui Jia
- Science and Experiment Research Center, Shenyang Medical College & Shenyang Key Laboratory of Vascular Biology, Science and Experimental Research Center, Shenyang Medical College, Shenyang, China
- School of Traditional Chinese Medicine, Shenyang Medical College, Shenyang, China
| | - Mei Feng
- Science and Experiment Research Center, Shenyang Medical College & Shenyang Key Laboratory of Vascular Biology, Science and Experimental Research Center, Shenyang Medical College, Shenyang, China
| | - Haolin Ren
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Junjia Gao
- Department of Cardiology, Second Affiliated Hospital, Shenyang Medical College, Shenyang, China
| | - Yueyang Liu
- Science and Experiment Research Center, Shenyang Medical College & Shenyang Key Laboratory of Vascular Biology, Science and Experimental Research Center, Shenyang Medical College, Shenyang, China
- School of Pharmacy, Shenyang Medical College, Shenyang, China
| | - Lu Zhang
- Science and Experiment Research Center, Shenyang Medical College & Shenyang Key Laboratory of Vascular Biology, Science and Experimental Research Center, Shenyang Medical College, Shenyang, China
| | - Ming-Sheng Zhou
- Science and Experiment Research Center, Shenyang Medical College & Shenyang Key Laboratory of Vascular Biology, Science and Experimental Research Center, Shenyang Medical College, Shenyang, China
| |
Collapse
|
21
|
Yang X, Liu C, Lei Y, Liu Z, Zhu B, Zhao D. PIM1 signaling in immunoinflammatory diseases: an emerging therapeutic target. Front Immunol 2024; 15:1443784. [PMID: 39372407 PMCID: PMC11449710 DOI: 10.3389/fimmu.2024.1443784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 09/02/2024] [Indexed: 10/08/2024] Open
Abstract
PIM1, the proviral integration site for Moloney murine leukemia virus, is a member of the serine/threonine protein kinase family. It is involved in many biological events, such as cell survival, cell cycle progression, cell proliferation, and cell migration, and has been widely studied in malignant diseases. However, recent studies have shown that PIM1 plays a prominent role in immunoinflammatory diseases, including autoimmune uveitis, inflammatory bowel disease, asthma, and rheumatoid arthritis. PIM1 can function in inflammatory signal transduction by phosphorylating multiple inflammatory protein substrates and mediating macrophage activation and T lymphocyte cell specification, thus participating in the development of multiple immunoinflammatory diseases. Moreover, the inhibition of PIM1 has been demonstrated to ameliorate certain immunoinflammatory disorders. Based on these studies, we suggest PIM1 as a potential therapeutic target for immunoinflammatory diseases and a valid candidate for future research. Herein, for the first time, we provide a detailed review that focuses on the roles of PIM1 in the pathogenesis of immunoinflammatory diseases.
Collapse
Affiliation(s)
- Xue Yang
- Department of Pediatrics, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, China
- Department of Pediatrics, Children’s Digital Health and Data Center, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Chunming Liu
- Department of Pediatrics, Children’s Digital Health and Data Center, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Yuxi Lei
- Department of Pediatrics, Children’s Digital Health and Data Center, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Zhi Liu
- Department of Pediatrics, Children’s Digital Health and Data Center, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Bin Zhu
- Department of Pediatrics, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, China
| | - Dongchi Zhao
- Department of Pediatrics, Children’s Digital Health and Data Center, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
22
|
Li S, Wang Y, Liu Q, Tang F, Zhang X, Yang S, Wang Q, Yang Q, Li S, Liu J, Han L, Liao Y, Yin X, Fan J, Feng H. RBC-hitchhiking PLGA nanoparticles loading β-glucan as a delivery system to enhance in vitro and in vivo immune responses in mice. Front Vet Sci 2024; 11:1462518. [PMID: 39351151 PMCID: PMC11439874 DOI: 10.3389/fvets.2024.1462518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 08/21/2024] [Indexed: 10/04/2024] Open
Abstract
Red blood cells (RBCs) naturally trap some bacterial pathogens in the circulation and kill them by oxidative stress. Following neutralization, the bacteria are presented to antigen-presenting cells in the spleen by the RBCs. This ability of RBCs has been harnessed to develop a system where they play a crucial role in enhancing the immune response, offering a novel approach to enhance the body's immunity. In this work, a conjugate, G-OVA, was formed by connecting β-glucan and OVA through a disulfide bond. Poly (lactic-co-glycolic acid) (PLGA) was then employed to encapsulate G-OVA, yielding G-OVA-PLGA. Finally, the nanoparticles were adsorbed onto RBCs to develop G-OVA-PLGA@RBC. The results demonstrated that the delivery of nanoparticles by RBCs enhanced the antibody response to antigens both in vitro and in vivo. The objective of this study was to investigate the increased immune activity of G-OVA-PLGA nanoparticles facilitated by RBCs transportation and to elucidate some of its underlying mechanisms. These findings are anticipated to contribute valuable insights for the development of efficient and safe immune enhancers.
Collapse
Affiliation(s)
- Sheng Li
- College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu, Sichuan, China
| | - Yao Wang
- College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu, Sichuan, China
| | - Qianqian Liu
- College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu, Sichuan, China
| | - Feng Tang
- College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu, Sichuan, China
| | - Xinnan Zhang
- College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu, Sichuan, China
| | - Shuyao Yang
- College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu, Sichuan, China
| | - Qiran Wang
- College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu, Sichuan, China
| | - Qian Yang
- College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu, Sichuan, China
| | - Shanshan Li
- Institute of Qinghai-Tibetan Plateau, Southwest Minzu University, Chengdu, Sichuan, China
| | - Jie Liu
- College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu, Sichuan, China
| | - Lu Han
- Institute of Qinghai-Tibetan Plateau, Southwest Minzu University, Chengdu, Sichuan, China
| | - Yi Liao
- College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu, Sichuan, China
| | - Xuemei Yin
- College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu, Sichuan, China
| | - Jing Fan
- College of Pharmacy, Chengdu University, Chengdu, China
| | - Haibo Feng
- College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu, Sichuan, China
| |
Collapse
|
23
|
Lv C, Li R, Yang D, Song S, Cheng X, Chen T, Chen L, Xiong Y. Broad-spectrum antiviral effect of MoringaA-loaded exosomes against IAV by mediating the GCN5-TFEB-autolysosome pathway. J Med Virol 2024; 96:e29906. [PMID: 39262090 DOI: 10.1002/jmv.29906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 08/02/2024] [Accepted: 08/23/2024] [Indexed: 09/13/2024]
Abstract
Influenza virus-induced viral pneumonia is a major threat to human health, and specific therapeutic agents for viral pneumonia are still lacking. MoringaA (MA) is an anti-influenza virus active compound isolated from Moringa seeds, which can inhibit influenza virus by activating the TFEB-autophagic lysosomal pathway in host cells. In this study, we obtained exosomes from M2-type macrophages and encapsulated and delivered MA (MA-Exos), and we investigated the efficacy of MA-Exos in antiviral and viral pneumonia in vivo and in vitro, respectively. In addition, we provided insights into the mechanism by which MA-Exos regulates TFEB-lysosomal autophagy by RNA sequencing. The MA-Exos showed broad-spectrum inhibition of IAV, and significant promotion of the autophagic lysosomal pathway. Meanwhile, we found that GCN5 gene and protein were significantly down-regulated in IAV-infected cells after MA-Exos intervention, indicating its blocking the acetylation of TFEB by GCN5. In addition, MA-Exos also significantly promoted autophagy in lung tissue cells of mice with viral pneumonia. MA-Exos can inhibit and clear influenza virus by mediating the TFEB-autophagy lysosomal pathway by a mechanism related to the down-regulation of histone acetyltransferase GCN5. Our study provides a strategy for targeting MA-Exos for the treatment of viral pneumonia from both antiviral and virus-induced inflammation inhibition pathways.
Collapse
Affiliation(s)
- Chunmei Lv
- Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou, China
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China
| | - Ruidong Li
- Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou, China
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China
| | - Dandan Yang
- Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou, China
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China
| | - Shunqiang Song
- Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou, China
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China
| | - Xu Cheng
- Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou, China
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China
| | - Tingting Chen
- Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou, China
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China
| | - Lei Chen
- Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou, China
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China
| | - Yongai Xiong
- Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou, China
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China
| |
Collapse
|
24
|
Zhou J, Ma X, Liu X, Liu Y, Fu J, Qi Y, Liu H. The impact of histone lactylation on the tumor microenvironment and metabolic pathways and its potential in cancer therapy. Genes Genomics 2024; 46:991-1011. [PMID: 39127851 DOI: 10.1007/s13258-024-01554-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 07/29/2024] [Indexed: 08/12/2024]
Abstract
BACKGROUND The complexity of cancer is intricately linked to its multifaceted biological processes, including the roles of the tumor microenvironment (TME) as well as genetic and metabolic regulation. Histone lactylation has recently emerged as a novel epigenetic modification mechanism that plays a pivotal role in regulating cancer initiation, proliferation, invasion, and metastasis. OBJECTIVE This review aims to elucidate the role of histone lactylation in modulating various aspects of tumor biology, including DNA repair mechanisms, glycolytic metabolic abnormalities, functions of non-tumor cells in the TME, and the promotion of tumor inflammatory responses and immune escape. Additionally, the review explores potential therapeutic strategies targeting histone lactylation. METHODS A comprehensive literature review was performed, analyzing recent findings on histone lactylation and its impact on cancer biology. This involved a systematic examination of studies focusing on biochemical pathways, cellular interactions, and clinical implications related to histone lactylation. RESULTS Histone lactylation was identified as a critical regulator of tumor cell DNA repair mechanisms and glycolytic metabolic abnormalities. It also significantly influences the functions of non-tumor cells within the TME, promoting tumor inflammatory responses and immune escape. Moreover, histone lactylation acts as a multifunctional biological signaling molecule impacting immune responses within the TME. Various cell types within the TME, including T cells and macrophages, were found to regulate tumor growth and immune escape mechanisms through lactylation. CONCLUSION Histone lactylation offers a novel perspective on tumor metabolism and its role in cancer development. It presents promising opportunities for the development of innovative cancer therapies. This review underscores the potential of histone lactylation as a therapeutic target, paving the way for new strategies in cancer treatment.
Collapse
Affiliation(s)
- Juanhong Zhou
- The First Clinical Medical College of Gansu University of Chinese Medicine, Gansu Provincial Hospital, Lanzhou, China
| | - Xinyun Ma
- The First Clinical Medical College of Gansu University of Chinese Medicine, Gansu Provincial Hospital, Lanzhou, China
| | - Xiaofeng Liu
- The First Clinical Medical College of Gansu University of Chinese Medicine, Gansu Provincial Hospital, Lanzhou, China
| | - Yang Liu
- The First Clinical Medical College of Gansu University of Chinese Medicine, Gansu Provincial Hospital, Lanzhou, China
| | - Jiaojiao Fu
- The First Clinical Medical College of Gansu University of Chinese Medicine, Gansu Provincial Hospital, Lanzhou, China
| | - Yaling Qi
- The First Clinical Medical College of Gansu University of Chinese Medicine, Gansu Provincial Hospital, Lanzhou, China
| | - Huiling Liu
- Department of Obstetrics and Gynecology, Gansu Provincial Hospital, Lanzhou, Gansu, China.
| |
Collapse
|
25
|
Ishibashi K, Hirata E. Multifaceted interactions between cancer cells and glial cells in brain metastasis. Cancer Sci 2024; 115:2871-2878. [PMID: 38992968 PMCID: PMC11462981 DOI: 10.1111/cas.16241] [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: 04/17/2024] [Revised: 05/20/2024] [Accepted: 05/26/2024] [Indexed: 07/13/2024] Open
Abstract
Cancer brain metastasis has a poor prognosis, is commonly observed in clinical practice, and the number of cases is increasing as overall cancer survival improves. However, experiments in mouse models have shown that brain metastasis itself is an inefficient process. One reason for this inefficiency is the brain microenvironment, which differs significantly from that of other organs, making it difficult for cancer cells to adapt. The brain microenvironment consists of unique resident cell types such as neurons, oligodendrocytes, astrocytes, and microglia. Accumulating evidence over the past decades suggests that the interactions between cancer cells and glial cells can positively or negatively influence the development of brain metastasis. Nevertheless, elucidating the complex interactions between cancer cells and glial cells remains challenging, in part due to the limitations of existing experimental models for glial cell culture. In this review, we first provide an overview of glial cell culture methods and then examine recent discoveries regarding the interactions between brain metastatic cancer cells and the surrounding glial cells, with a special focus on astrocytes and microglia. Finally, we discuss future perspectives for understanding the multifaceted interactions between cancer cells and glial cells for the treatment of metastatic brain tumors.
Collapse
Affiliation(s)
- Kojiro Ishibashi
- Division of Tumor Cell Biology and BioimagingCancer Research Institute of Kanazawa UniversityKanazawaIshikawaJapan
| | - Eishu Hirata
- Division of Tumor Cell Biology and BioimagingCancer Research Institute of Kanazawa UniversityKanazawaIshikawaJapan
- WPI Nano Life Science Institute, Kanazawa UniversityKanazawaIshikawaJapan
| |
Collapse
|
26
|
Wu H, Zhang L, Dong X, Yang J, Zheng L, Li L, Liu X, Jin M, Zhang P. Targeted delivery of berberine using bionic nanomaterials for Atherosclerosis therapy. Biomed Pharmacother 2024; 178:117135. [PMID: 39047421 DOI: 10.1016/j.biopha.2024.117135] [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: 05/06/2024] [Revised: 07/07/2024] [Accepted: 07/10/2024] [Indexed: 07/27/2024] Open
Abstract
Atherosclerosis (AS) is a prevalent chronic vascular inflammatory disease globally, initiated by injury to vascular endothelial cells (VECs). Macrophages play a pivotal role in disease pathogenesis, involving lipid metabolism and inflammation. The application of nanomaterials has been hindered by their rapid clearance by the immune system. Utilizing macrophage cell membranes can mitigate abnormal immune responses and induce a "homing" effect. Here, M2 macrophage cell membranes (M2) were coated onto berberine polylactic-hydroxylase-polylactide (PLGA) nanoparticles (BBR NPs), employing M2 macrophage immune escape, "homing" ability, and membrane coating nanotechnology, and loaded with mannose (Man) to create bionic nanoparticles (BBR NPs@Man/M2). Subsequently, the physical properties of BBR NPs@Man/M2 were characterized. The biocompatibility and biological function of BBR NPs@Man/M2 were assessed in vitro. Finally, the targeting, therapeutic efficacy, and safety of BBR NPs@M2 were investigated in an AS mouse model. The newly developed BBR NPs@Man/M2 exhibited good biocompatibility. Owing to their M2 coating, the nanoparticles effectively targeted macrophages in vitro, inducing a shift from a pro-inflammatory to an anti-inflammatory state. This transition reduced inflammation in endothelial cells and facilitated the repair of damaged endothelial cells. Moreover, M2-coated nanoparticles efficiently targeted and accumulated in atherosclerotic lesions in vivo. Following four weeks of treatment, BBR NPs@Man/M2 significantly delayed AS progression. Furthermore, BBR NPs@Man/M2 demonstrated a good safety profile after long-term administration. In conclusion, BBR NPs@Man/M2 effectively and safely inhibited AS progression. Biomimetic nanoparticles represent a promising approach for the safe and effective delivery of anti-AS drugs.
Collapse
Affiliation(s)
- Haopeng Wu
- Department of Cardiovascular, Affifiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Lei Zhang
- Department of Cardiovascular, Affifiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Xiaokang Dong
- Department of Cardiovascular, Affifiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Jie Yang
- Department of Cardiovascular, Affifiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Liang Zheng
- Department of Cardiovascular, Affifiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Lei Li
- Department of Cardiovascular, Affifiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Xiujuan Liu
- Department of Cardiovascular, Affifiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Meiying Jin
- Yantai Affiliated Hospital of Binzhou Medical College, Yantai 264000, China
| | - Peng Zhang
- College of traditional Chinese medicine, Binzhou Medical University, Yantai 264000, China.
| |
Collapse
|
27
|
Lan M, Lin C, Zeng L, Hu S, Shi Y, Zhao Y, Liu X, Sun J, Liang G, Huang M. Linderanine C regulates macrophage polarization by inhibiting the MAPK signaling pathway against ulcerative colitis. Biomed Pharmacother 2024; 178:117239. [PMID: 39098180 DOI: 10.1016/j.biopha.2024.117239] [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: 06/03/2024] [Revised: 07/26/2024] [Accepted: 07/30/2024] [Indexed: 08/06/2024] Open
Abstract
Ulcerative colitis (UC) is a chronic non-specific inflammatory disease involving the mucosa and submucosa of the rectum and colon. Lindera aggregate (Sims) Kosterm is a traditional Chinese herb used for thousands of years in the treatment of gastrointestinal diseases. Previously, we have demonstrated that the extracts of Lindera aggregate have good anti-UC effects, but their pharmacodynamic active components have not been fully clarified. Therefore, we explored the therapeutic effect of Linderanine C (LDC), a characteristic component of Lindera aggregata, on UC and its mechanism in this study. Firstly, we found that LDC could significantly reduce the disease activity index of UC and improve shortened colon and pathological changes in vivo. Colon tissue transcriptomics suggested that the anti-UC effect of LDC might be related to its anti-inflammatory activity. Cellular experiments revealed that LDC could inhibit the expression of the M1 cell marker CD86 in RAW264.7 cells, reduce the production of inflammatory mediators such as IL-6 and TNF-α, and have good anti-inflammatory activity in vitro. Cellular transcriptomics reveal the potential involvement of the MAPK signaling pathway in the anti-inflammatory effect of LDC. The co-culture assay confirmed that LDC could significantly reduce inflammation-mediated intestinal epithelial cell injury. In conclusion, LDC was able to inhibit macrophage M1 polarization and reduce inflammatory mediator production by inhibiting the MAPK signaling pathway, effectively improving UC.
Collapse
Affiliation(s)
- Mengyao Lan
- Zhejiang TCM Key Laboratory of Pharmacology and Translational Research of Natural Products, School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang 311399, China
| | - Cailu Lin
- Zhejiang TCM Key Laboratory of Pharmacology and Translational Research of Natural Products, School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang 311399, China
| | - Lulu Zeng
- Zhejiang TCM Key Laboratory of Pharmacology and Translational Research of Natural Products, School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang 311399, China
| | - Shijie Hu
- Zhejiang TCM Key Laboratory of Pharmacology and Translational Research of Natural Products, School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang 311399, China
| | - Yuan Shi
- Zhejiang TCM Key Laboratory of Pharmacology and Translational Research of Natural Products, School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang 311399, China
| | - Yan Zhao
- Zhejiang TCM Key Laboratory of Pharmacology and Translational Research of Natural Products, School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang 311399, China
| | - Xin Liu
- Department of Colorectal Surgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| | - Jinfeng Sun
- Department of Pharmacy and Institute of Inflammation, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, China; Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, Jilin 133002, China
| | - Guang Liang
- Zhejiang TCM Key Laboratory of Pharmacology and Translational Research of Natural Products, School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang 311399, China; Department of Pharmacy and Institute of Inflammation, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, China.
| | - Mincong Huang
- Zhejiang TCM Key Laboratory of Pharmacology and Translational Research of Natural Products, School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang 311399, China.
| |
Collapse
|
28
|
Li A, Li Y, Li X, Tang C, Yang Y, Li N, Jin Y. Ferritin light chain as a potential biomarker for the prognosis of liver hepatocellular carcinoma. Heliyon 2024; 10:e36040. [PMID: 39224384 PMCID: PMC11367121 DOI: 10.1016/j.heliyon.2024.e36040] [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: 03/27/2024] [Revised: 07/11/2024] [Accepted: 08/08/2024] [Indexed: 09/04/2024] Open
Abstract
High expression of the ferritin light chain (FTL) in cancer promotes its onset and progression and is associated with tumour evolution. However, the significance of FTL in pan-cancer progression and prognosis in humans remains unclear. Therefore, we selected various bioinformatics databases to perform a pan-cancer analysis on a public dataset. Our results showed that FTL was differentially expressed in pan-cancer tissues compared to normal tissues. High FTL expression significantly correlated with the clinicopathological characteristics of patients with liver hepatocellular carcinoma (LIHC). The subsequent validation experiments confirmed these observations. Notably, our study found for the first time that FTLs are closely associated with LIHC and that FTLs have important clinical diagnostic and prognostic value for patients with LIHC. We confirmed that FTL expression was closely associated with altered DNA cycles and immune infiltration in LIHC. In conclusion, high levels of FTL expression are associated with poor prognosis in LIHC patients and are expected to be a potential prognostic and immune marker for LIHC.
Collapse
Affiliation(s)
- Aoqun Li
- Central Laboratory, The Affiliated Hospital of Yanbian University, Yanji, 133000, China
- Key Laboratory of Tumor Pathobiology (Yanbian University), State Ethnic Affairs Commission, Yanji, 133000, China
| | - Yue Li
- Central Laboratory, The Affiliated Hospital of Yanbian University, Yanji, 133000, China
- Key Laboratory of Tumor Pathobiology (Yanbian University), State Ethnic Affairs Commission, Yanji, 133000, China
| | - Xiaoqing Li
- Central Laboratory, The Affiliated Hospital of Yanbian University, Yanji, 133000, China
- Key Laboratory of Tumor Pathobiology (Yanbian University), State Ethnic Affairs Commission, Yanji, 133000, China
| | - Chunxiao Tang
- Central Laboratory, The Affiliated Hospital of Yanbian University, Yanji, 133000, China
- Key Laboratory of Tumor Pathobiology (Yanbian University), State Ethnic Affairs Commission, Yanji, 133000, China
| | - Yang Yang
- Central Laboratory, The Affiliated Hospital of Yanbian University, Yanji, 133000, China
- Key Laboratory of Tumor Pathobiology (Yanbian University), State Ethnic Affairs Commission, Yanji, 133000, China
| | - Nan Li
- Institute of Virology, Wenzhou University, Wenzhou, 325000, China
| | - Yun Jin
- Central Laboratory, The Affiliated Hospital of Yanbian University, Yanji, 133000, China
- Department of Ultrasound, The Affiliated Hospital of Yanbian University, Yanji, 133000, China
| |
Collapse
|
29
|
Won EJ, Lee M, Lee EK, Baek SH, Yoon TJ. Lipid-Based Nanoparticles Fused with Natural Killer Cell Plasma Membrane Proteins for Triple-Negative Breast Cancer Therapy. Pharmaceutics 2024; 16:1142. [PMID: 39339179 PMCID: PMC11434974 DOI: 10.3390/pharmaceutics16091142] [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: 07/29/2024] [Revised: 08/14/2024] [Accepted: 08/26/2024] [Indexed: 09/30/2024] Open
Abstract
Immunotherapy combined with chemicals and genetic engineering tools is emerging as a promising strategy to treat triple-negative breast cancer (TNBC), which is more aggressive with poorer progress than other breast cancer subtypes. In this study, lipid-based nanoparticles (LNPs) possessed an NK cell-like function that could deliver tumor-specific therapeutics and inhibit tumor growth. LNPs fused with an NK cell membrane protein system (NK-LNP) have three main features: (i) hydrophilic plasmid DNA can inhibit TNBC metastasis when encapsulated within LNPs and delivered to cells; (ii) the lipid composition of LNPs, including C18 ceramide, exhibits anticancer effects; (iii) NK cell membrane proteins are immobilized on the LNP surface, enabling targeted delivery to TNBC cells. These particles facilitate the targeted delivery of HIC1 plasmid DNA and the modulation of immune cell functions. Delivered therapeutic genes can inhibit metastasis of TNBC and then induce apoptotic cell death while targeting macrophages to promote cytokine release. The anticancer effect is expected to be applied in treating various difficult-to-treat cancers with LNP fused with NK cell plasma membrane proteins, which can simultaneously deliver therapeutic chemicals and genes.
Collapse
Affiliation(s)
- Eun-Jeong Won
- Research Institute of Pharmaceutical Science and Technology (RIPST), Department of Pharmacy, Ajou University, 206 Worldcup-ro, Yeongtong-gu, Suwon 16499, Republic of Korea
- Nucleic Acid Therapeutics Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 30 Yeongudanji-ro, Ochang, Cheongwon, Cheongju 28116, Republic of Korea
| | - Myungchul Lee
- School of Pharmacy, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon 31065, Republic of Korea
| | - Eui-Kyung Lee
- School of Pharmacy, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon 31065, Republic of Korea
| | - Seung-Hoon Baek
- Research Institute of Pharmaceutical Science and Technology (RIPST), Department of Pharmacy, Ajou University, 206 Worldcup-ro, Yeongtong-gu, Suwon 16499, Republic of Korea
| | - Tae-Jong Yoon
- Research Institute of Pharmaceutical Science and Technology (RIPST), Department of Pharmacy, Ajou University, 206 Worldcup-ro, Yeongtong-gu, Suwon 16499, Republic of Korea
- Department of BioHealth Regulatory Science, Graduate School of Ajou University, 206 Worldcup-ro, Yeongtong-gu, Suwon 16499, Republic of Korea
- Moogene Medi Institute, 25, Misagangbyeonjungang-ro 7beonan-gil, Hanam 12939, Republic of Korea
| |
Collapse
|
30
|
Qu Z, Luo J, Li Z, Yang R, Zhao J, Chen X, Yu S, Shu H. Advancements in strategies for overcoming the blood-brain barrier to deliver brain-targeted drugs. Front Aging Neurosci 2024; 16:1353003. [PMID: 39253614 PMCID: PMC11381257 DOI: 10.3389/fnagi.2024.1353003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Accepted: 08/06/2024] [Indexed: 09/11/2024] Open
Abstract
The blood-brain barrier is known to consist of a variety of cells and complex inter-cellular junctions that protect the vulnerable brain from neurotoxic compounds; however, it also complicates the pharmacological treatment of central nervous system disorders as most drugs are unable to penetrate the blood-brain barrier on the basis of their own structural properties. This dramatically diminished the therapeutic effect of the drug and compromised its biosafety. In response, a number of drugs are often delivered to brain lesions in invasive ways that bypass the obstruction of the blood-brain barrier, such as subdural administration, intrathecal administration, and convection-enhanced delivery. Nevertheless, these intrusive strategies introduce the risk of brain injury, limiting their clinical application. In recent years, the intensive development of nanomaterials science and the interdisciplinary convergence of medical engineering have brought light to the penetration of the blood-brain barrier for brain-targeted drugs. In this paper, we extensively discuss the limitations of the blood-brain barrier on drug delivery and non-invasive brain-targeted strategies such as nanomedicine and blood-brain barrier disruption. In the meantime, we analyze their strengths and limitations and provide outlooks on the further development of brain-targeted drug delivery systems.
Collapse
Affiliation(s)
- Zhichuang Qu
- Department of Neurosurgery, Meishan City People's Hospital, Meishan, China
- Department of Neurosurgery, General Hospital of Western Theater Command, Chengdu, China
| | - Juan Luo
- Department of Neurosurgery, General Hospital of Western Theater Command, Chengdu, China
- Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Zheng Li
- Department of Neurosurgery, General Hospital of Western Theater Command, Chengdu, China
- Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Rong Yang
- Department of Neurosurgery, General Hospital of Western Theater Command, Chengdu, China
- Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jiaxi Zhao
- Department of Neurosurgery, General Hospital of Western Theater Command, Chengdu, China
- Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Xin Chen
- Department of Neurosurgery, General Hospital of Western Theater Command, Chengdu, China
| | - Sixun Yu
- Department of Neurosurgery, General Hospital of Western Theater Command, Chengdu, China
- College of Medicine of Southwest Jiaotong University, Chengdu, China
| | - Haifeng Shu
- Department of Neurosurgery, General Hospital of Western Theater Command, Chengdu, China
- College of Medicine of Southwest Jiaotong University, Chengdu, China
| |
Collapse
|
31
|
Xian Y, Liu J, Dai M, Zhang W, He M, Wei Z, Jiang Y, Le S, Lin Z, Tang S, Zhou Y, Dong L, Liang J, Zhang J, Wang L. Microglia Promote Lymphangiogenesis Around the Spinal Cord Through VEGF-C/VEGFR3-Dependent Autophagy and Polarization After Acute Spinal Cord Injury. Mol Neurobiol 2024:10.1007/s12035-024-04437-5. [PMID: 39158788 DOI: 10.1007/s12035-024-04437-5] [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: 01/25/2024] [Accepted: 08/09/2024] [Indexed: 08/20/2024]
Abstract
Reducing secondary injury is a key focus in the field of spinal cord injury (SCI). Recent studies have revealed the role of lymphangiogenesis in reducing secondary damage to central nerve. However, the mechanism of lymphangiogenesis is not yet clear. Macrophages have been shown to play an important role in peripheral tissue lymphangiogenesis. Microglia is believed to play a role similar to macrophages in the central nervous system (CNS); we hypothesized that there was a close relationship between microglia and central nerve system lymphangiogenesis. Herein, we used an in vivo model of SCI to explored the relationship between microglia and spinal cord lymphangiogenesis and further investigated the polarization of microglia and its role in promoting spinal cord lymphangiogenesis by a series of in vitro experiments. The current study elucidated for the first time the relationship between microglia and lymphangiogenesis around the spinal cord after SCI. Classical activated (M1) microglia can promote lymphangiogenesis by secreting VEGF-C which further increases polarization and secretion of lymphatic growth factor by activating VEGFR3. The VEGF-C/VEGFR3 pathway activation downregulates microglia autophagy, thereby regulating the microglia phenotype. These results indicate that M1 microglia promote lymphangiogenesis after SCI, and activated VEGF-C/VEGFR3 signaling promotes M1 microglia polarization by inhibiting autophagy, thereby facilitates lymphangiogenesis.
Collapse
Grants
- 202102020768 Science and Technology Program of Guangzhou, China
- 202102020768 Science and Technology Program of Guangzhou, China
- 202102020768 Science and Technology Program of Guangzhou, China
- 202102020768 Science and Technology Program of Guangzhou, China
- 202102020768 Science and Technology Program of Guangzhou, China
- 202102020768 Science and Technology Program of Guangzhou, China
- 202102020768 Science and Technology Program of Guangzhou, China
- 202102020768 Science and Technology Program of Guangzhou, China
- 202102020768 Science and Technology Program of Guangzhou, China
- 202102020768 Science and Technology Program of Guangzhou, China
- 202102020768 Science and Technology Program of Guangzhou, China
- 202102020768 Science and Technology Program of Guangzhou, China
- 202102020768 Science and Technology Program of Guangzhou, China
- 202102020768 Science and Technology Program of Guangzhou, China
- 202102020768 Science and Technology Program of Guangzhou, China
- 82072433 National Natural Science Foundation of China
- 82072433 National Natural Science Foundation of China
- 82072433 National Natural Science Foundation of China
- 82072433 National Natural Science Foundation of China
- 82072433 National Natural Science Foundation of China
- 82072433 National Natural Science Foundation of China
- 82072433 National Natural Science Foundation of China
- 82072433 National Natural Science Foundation of China
- 82072433 National Natural Science Foundation of China
- 82072433 National Natural Science Foundation of China
- 82072433 National Natural Science Foundation of China
- 82072433 National Natural Science Foundation of China
- 82072433 National Natural Science Foundation of China
- 82072433 National Natural Science Foundation of China
- 82072433 National Natural Science Foundation of China
- 2214050002081 Natural Science Foundation of Guangdong Province
- 2214050002081 Natural Science Foundation of Guangdong Province
- 2214050002081 Natural Science Foundation of Guangdong Province
- 2214050002081 Natural Science Foundation of Guangdong Province
- 2214050002081 Natural Science Foundation of Guangdong Province
- 2214050002081 Natural Science Foundation of Guangdong Province
- 2214050002081 Natural Science Foundation of Guangdong Province
- 2214050002081 Natural Science Foundation of Guangdong Province
- 2214050002081 Natural Science Foundation of Guangdong Province
- 2214050002081 Natural Science Foundation of Guangdong Province
- 2214050002081 Natural Science Foundation of Guangdong Province
- 2214050002081 Natural Science Foundation of Guangdong Province
- 2214050002081 Natural Science Foundation of Guangdong Province
- 2214050002081 Natural Science Foundation of Guangdong Province
- 2214050002081 Natural Science Foundation of Guangdong Province
Collapse
Affiliation(s)
- Yeyang Xian
- Tianhe District, Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Zhongshandadao West 183, Guangzhou City, 510000, China
| | - Jie Liu
- Tianhe District, Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Zhongshandadao West 183, Guangzhou City, 510000, China
| | - Mengxuan Dai
- Tianhe District, Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Zhongshandadao West 183, Guangzhou City, 510000, China
| | - Wensheng Zhang
- Tianhe District, Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Zhongshandadao West 183, Guangzhou City, 510000, China
| | - Minye He
- Tianhe District, Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Zhongshandadao West 183, Guangzhou City, 510000, China
| | - Zhengnong Wei
- Tianhe District, Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Zhongshandadao West 183, Guangzhou City, 510000, China
| | - Yutao Jiang
- Tianhe District, Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Zhongshandadao West 183, Guangzhou City, 510000, China
| | - Shiyong Le
- Tianhe District, Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Zhongshandadao West 183, Guangzhou City, 510000, China
| | - Zhuoang Lin
- Tianhe District, Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Zhongshandadao West 183, Guangzhou City, 510000, China
| | - Shuai Tang
- Tianhe District, Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Zhongshandadao West 183, Guangzhou City, 510000, China
| | - Yunfei Zhou
- Tianhe District, Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Zhongshandadao West 183, Guangzhou City, 510000, China
| | - Liming Dong
- Tianhe District, Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Zhongshandadao West 183, Guangzhou City, 510000, China
| | - Jinzheng Liang
- Tianhe District, Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Zhongshandadao West 183, Guangzhou City, 510000, China
| | - Jie Zhang
- Tianhe District, Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Zhongshandadao West 183, Guangzhou City, 510000, China.
| | - Liang Wang
- Tianhe District, Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Zhongshandadao West 183, Guangzhou City, 510000, China.
| |
Collapse
|
32
|
Huang Z, Shen X, Chen H, Zhu Z, Lu B, Zhang L, Liu Y, Li Y, Xu B. Zhiqiao Gancao decoction regulated JAK2/STAT3/ macrophage M1 polarization to ameliorate intervertebral disc degeneration. Heliyon 2024; 10:e34715. [PMID: 39170327 PMCID: PMC11336307 DOI: 10.1016/j.heliyon.2024.e34715] [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: 02/26/2024] [Revised: 07/13/2024] [Accepted: 07/15/2024] [Indexed: 08/23/2024] Open
Abstract
Background Zhiqiao Gancao decoction (ZQGCD) was created by Professor Gong Zhengfeng, a renowned Chinese medicine expert. Clinical studies have shown its efficacy in alleviating pain and enhancing lumbar function in intervertebral disc degeneration (IDD) patients. However, the precise mechanism of ZQGCD in treating IDD remains unclear. Methods The active components of ZQGCD were identified using Liquid chromatography-tandem mass spectrometry (LC-MS/MS). A rat model of intervertebral disc degeneration was established, and rats in each group received ZQGCD for three weeks. Assessment parameters included hyperalgesia status, observation of intervertebral disc tissue degeneration and macrophage infiltration, and analysis of JAK2/STAT3 pathway protein expression in the intervertebral disc. Primary macrophage M1 polarization was induced using LPS, with cells treated using the JAK2 inhibitor (AZD1480) and ZQGCD to evaluate macrophage polarization, cellular supernatant inflammatory factors, and JAK2/STAT3 pathway expression. Macrophage supernatant served as a conditioned medium to observe its effects on the proliferation of nucleus pulposus cells (NPCs) and the expression of collagen II and MMP3 proteins. Results A total of 81 active components were identified in ZQGCD. Following ZQGCD treatment, infiltrating macrophages in intervertebral disc tissues of model rats decreased, the content of M1 macrophages decreased, while the content of M2 macrophages increased, the expression of proinflammatory factors and pain-inducing factors in serum decreased, and the expression of substance P in intervertebral disc tissue decreased. Consequently, the intervertebral disc degeneration and hyperalgesia of rats were improved. In vitro studies revealed that LPS induced M1 macrophage polarization. By inhibiting the JAK2/STAT3 pathway, both JAK2 inhibitors and ZQGCD effectively suppressed M1 polarization, resulting in decreased levels of IL-1β, IL-6, TNF-α, and various other inflammatory factors. Consequently, this inhibition led to a delay in the degeneration of NPCs. Conclusion There is macrophage infiltration in the intervertebral disc tissue of IDD rats, and JAK2/STAT3 pathway is activated, macrophages are polarized to M1 type, resulting in inflammatory microenvironment, leading to intervertebral disc degeneration and hyperalgesia. ZQGCD exhibited a delaying effect on IDD and improved hyperalgesia by inhibiting the JAK2/STAT3/macrophage M1 polarization pathway.
Collapse
Affiliation(s)
- Zeling Huang
- Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, Jiangsu, 215009, China
| | - Xiaofeng Shen
- Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, Jiangsu, 215009, China
- Orthopaedic Traumatology Institute, Suzhou Academy of Wumen Chinese Medicine, Suzhou, Jiangsu, 215009, China
| | - Hua Chen
- Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, Jiangsu, 215009, China
| | - Zaishi Zhu
- Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, Jiangsu, 215009, China
| | - Binjie Lu
- Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, Jiangsu, 215009, China
| | - Long Zhang
- Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, Jiangsu, 215009, China
| | - Yujiang Liu
- Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, Jiangsu, 215009, China
| | - Yuwei Li
- Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, Jiangsu, 215009, China
- Orthopaedic Traumatology Institute, Suzhou Academy of Wumen Chinese Medicine, Suzhou, Jiangsu, 215009, China
| | - Bo Xu
- Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, Jiangsu, 215009, China
| |
Collapse
|
33
|
Sayson SG, Ashbaugh A, Porollo A, Smulian G, Cushion MT. Pneumocystis murina promotes inflammasome formation and NETosis during Pneumocystis pneumonia. mBio 2024; 15:e0140924. [PMID: 38953359 PMCID: PMC11323544 DOI: 10.1128/mbio.01409-24] [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: 05/09/2024] [Accepted: 06/04/2024] [Indexed: 07/04/2024] Open
Abstract
Pneumocystis jirovecii pneumonia (PjP) poses a serious risk to individuals with compromised immune systems, such as individuals with HIV/AIDS or undergoing immunosuppressive therapies for cancer or solid organ transplants. Severe PjP triggers excessive lung inflammation, resulting in lung function decline and consequential alveolar damage, potentially culminating in acute respiratory distress syndrome. Non-HIV patients face a 30%-60% mortality rate, emphasizing the need for a deeper understanding of inflammatory responses in PjP. Prior research emphasized macrophages in Pneumocystis infections, neglecting neutrophils' role in tissue damage. Consequently, the overemphasis on macrophages led to an incomplete understanding of the role of neutrophils and inflammatory responses. In the current investigation, our RNAseq studies on a murine surrogate model of PjP revealed heightened activation of the NLRP3 inflammasome and NETosis cell death pathways in their lungs. Immunofluorescence staining confirmed neutrophil extracellular trap (NET) presence in the lungs of the P. murina-infected mice, validating our findings. Moreover, isolated neutrophils exhibited NETosis when directly stimulated with P. murina. Isolated NETs compromised P. murina viability in vitro, highlighting the potential role of neutrophils in controlling fungal growth and promoting inflammation during P. murina pneumonia through NLRP3 inflammasome assembly and NETosis. These pathways, essential for inflammation and pathogen elimination, bear the risk of uncontrolled activation leading to excessive tissue damage and persistent inflammation. This pioneering study is the first to identify the formation of NETs and inflammasomes during Pneumocystis infection, paving the way for comprehensive investigations into treatments aimed at mitigating lung damage and augmenting survival rates for individuals with PjP.IMPORTANCEPneumocystis jirovecii pneumonia (PjP) affects individuals with weakened immunity, such as HIV/AIDS, cancer, and organ transplant patients. Severe PjP triggers lung inflammation, impairing function and potentially causing acute respiratory distress syndrome. Non-HIV individuals face a 30%-60% mortality rate, underscoring the need for deeper insight into PjP's inflammatory responses. Past research focused on macrophages in managing Pneumocystis infection and its inflammation, while the role of neutrophils was generally overlooked. In contrast, our findings in P. murina-infected mouse lungs showed neutrophil involvement during inflammation and increased expression of NLRP3 inflammasome and NETosis pathways. Detection of neutrophil extracellular traps further indicated their involvement in the inflammatory process. Although beneficial in combating infection, unregulated neutrophil activation poses a potential threat to lung tissues. Understanding the behavior of neutrophils in Pneumocystis infections is crucial for controlling detrimental reactions and formulating treatments to reduce lung damage, ultimately improving the survival rates of individuals with PjP.
Collapse
Affiliation(s)
- Steven G. Sayson
- Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- The Veterans Affairs Medical Center, Cincinnati, Ohio, USA
| | - Alan Ashbaugh
- Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- The Veterans Affairs Medical Center, Cincinnati, Ohio, USA
| | - Aleksey Porollo
- Division of Human Genetics, Center for Autoimmune Genomics and Etiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, USA
| | - George Smulian
- Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- The Veterans Affairs Medical Center, Cincinnati, Ohio, USA
| | - Melanie T. Cushion
- Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- The Veterans Affairs Medical Center, Cincinnati, Ohio, USA
| |
Collapse
|
34
|
Chambers KL, Myerscough MR, Watson MG, Byrne HM. Blood Lipoproteins Shape the Phenotype and Lipid Content of Early Atherosclerotic Lesion Macrophages: A Dual-Structured Mathematical Model. Bull Math Biol 2024; 86:112. [PMID: 39093509 PMCID: PMC11297092 DOI: 10.1007/s11538-024-01342-9] [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: 04/10/2024] [Accepted: 07/16/2024] [Indexed: 08/04/2024]
Abstract
Macrophages in atherosclerotic lesions exhibit a spectrum of behaviours or phenotypes. The phenotypic distribution of monocyte-derived macrophages (MDMs), its correlation with MDM lipid content, and relation to blood lipoprotein densities are not well understood. Of particular interest is the balance between low density lipoproteins (LDL) and high density lipoproteins (HDL), which carry bad and good cholesterol respectively. To address these issues, we have developed a mathematical model for early atherosclerosis in which the MDM population is structured by phenotype and lipid content. The model admits a simpler, closed subsystem whose analysis shows how lesion composition becomes more pathological as the blood density of LDL increases relative to the HDL capacity. We use asymptotic analysis to derive a power-law relationship between MDM phenotype and lipid content at steady-state. This relationship enables us to understand why, for example, lipid-laden MDMs have a more inflammatory phenotype than lipid-poor MDMs when blood LDL lipid density greatly exceeds HDL capacity. We show further that the MDM phenotype distribution always attains a local maximum, while the lipid content distribution may be unimodal, adopt a quasi-uniform profile or decrease monotonically. Pathological lesions exhibit a local maximum in both the phenotype and lipid content MDM distributions, with the maximum at an inflammatory phenotype and near the lipid content capacity respectively. These results illustrate how macrophage heterogeneity arises in early atherosclerosis and provide a framework for future model validation through comparison with single-cell RNA sequencing data.
Collapse
Affiliation(s)
- Keith L Chambers
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Andrew Wiles Building, Radcliffe Observatory Quarter, Woodstock Road, Oxford, Oxfordshire, OX2 6GG, UK.
| | - Mary R Myerscough
- School of Mathematics and Statistics, University of Sydney, Carslaw Building, Eastern Avenue, Camperdown, Sydney, NSW, 2006, Australia
| | - Michael G Watson
- School of Mathematics and Statistics, University of New South Wales, Anita B. Lawrence Centre, University Mall, UNSW, Kensington, Sydney, NSW, 2052, Australia
| | - Helen M Byrne
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Andrew Wiles Building, Radcliffe Observatory Quarter, Woodstock Road, Oxford, Oxfordshire, OX2 6GG, UK
- Ludwig Institute for Cancer Research, University of Oxford, Old Road Campus Research Build, Roosevelt Dr, Headington, Oxford, Oxfordshire, OX3 7DQ, UK
| |
Collapse
|
35
|
Pei J, Zhang J, Yu C, Luo J, Wen S, Hua Y, Wei G. Transcriptomics-based exploration of shared M1-type macrophage-related biomarker in acute kidney injury after kidney transplantation and acute rejection after kidney transplantation. Transpl Immunol 2024; 85:102066. [PMID: 38815767 DOI: 10.1016/j.trim.2024.102066] [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: 12/23/2023] [Revised: 05/12/2024] [Accepted: 05/27/2024] [Indexed: 06/01/2024]
Abstract
BACKGROUND Macrophage type 1 (M1) cells are associated with both acute kidney injury (AKI) during kidney transplantation and acute rejection (AR) after kidney transplantation. Our study explored M1-related biomarkers involved in both AKI and AR and their potential biological functions. METHODS Based on the Gene Expression Omnibus (GEO) database, the immune cell infiltration levels and differentially expressed genes were examined in AKI and AR in the kidney transplantation; M1-related genes shared in AKI and AR were identified using weighted gene co-expression analysis (WGCNA) system. Subsequently, protein-protein interaction (PPI) networks and machine learning methods to identify Hub genes and construct diagnostic models. Both AKI model and AR rat models were built to validate the expressions of Hub genes and test the injury phenotype, oxidative stress markers, and inflammatory factors. Finally, the transcription factor (TF)-Hub gene and micro-RNA (miRNA)-Hub gene regulatory networks were constructed based on identified Hub genes. RESULTS Out of 2167 differential expression genes (DEGs) in AKI and 2100 DEGs in AR, four M1-related Hub genes were obtained by PPI networks and machine learning methods, namely GBP2, TYROBP, CCR5, and TLR8. The calibration curves in the nomogram diagnostic model for these four Hub genes suggested the same predictive probability as an ideal model for AKI and AR after kidney transplantation (AUC values of the area under the ROC curve were all >0.7). The same observations were confirmed in ischemia reperfusion injury (IRI) and AR rat models by identifying common four Hub genes (GBP2, TYROBP, TLR8, and CCR5). Western blots showed that these four Hub genes were significantly different in rat models of IRI and AR (all p<0.05). Compared with the control group, IRI and AR groups showed aggravated histopathological damage and increased secretion of oxidative stress markers and inflammatory factors in rat kidneys (all p<0.05). Finally, TF-Hub and miRNA-Hub gene regulatory networks were constructed to provide a theoretical basis for the regulation of Hub genes. CONCLUSION We identified four macrophage M1-related Hub genes shared among AKI and AR after kidney transplantation. These genes may be considered for diagnosis of AKI and AR after kidney transplantation.
Collapse
Affiliation(s)
- Jun Pei
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, China; Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, China
| | - Jie Zhang
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, China; Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, China
| | - Chengjun Yu
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, China; Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, China
| | - Jin Luo
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, China; Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, China
| | - Sheng Wen
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, China; Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, China
| | - Yi Hua
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, China; Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, China.
| | - Guanghui Wei
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, China; Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, China.
| |
Collapse
|
36
|
Li D, Gao S. The interplay between T lymphocytes and macrophages in myocardial ischemia/reperfusion injury. Mol Cell Biochem 2024; 479:1925-1936. [PMID: 37540399 DOI: 10.1007/s11010-023-04822-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 07/24/2023] [Indexed: 08/05/2023]
Abstract
Acute myocardial infarction is one of the most important causes of death in the world, causing a huge health and economic burden to the world. It is still a ticklish problem how to effectively prevent reperfusion injury while recovering the blood flow of ischemic myocardium. During the process of myocardial ischemia/reperfusion injury (MI/RI), the modulation of immune cells plays an important role. Monocyte/macrophage, neutrophils and endothelial cells initiate the inflammatory response and induce the release of various inflammatory cytokines, resulting in increased vascular permeability, tissue edema and damage. Meanwhile, T cells were recruited to impaired myocardium and release pro-inflammatory and anti-inflammatory cytokines. T cells and macrophages play important roles in keeping cardiac homeostasis and orchestrate tissue repair. T cells differentiation and macrophages polarization precisely regulates the tissue microenvironment in MI/RI, and shows cross action, but the mechanism is unclear. To identify potential intervention targets and propose ideas for treatment and prevention of MI/RI, this review explores the crosstalk between T lymphocytes and macrophages in MI/RI.
Collapse
Affiliation(s)
- Dan Li
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, 314 An Shan Xi Road, Nan Kai District, Tianjin, 300193, China
- Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, China
| | - Shan Gao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, 314 An Shan Xi Road, Nan Kai District, Tianjin, 300193, China.
- Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, China.
| |
Collapse
|
37
|
Ganguly K, Luthfikasari R, Randhawa A, Dutta SD, Patil TV, Acharya R, Lim KT. Stimuli-Mediated Macrophage Switching, Unraveling the Dynamics at the Nanoplatforms-Macrophage Interface. Adv Healthc Mater 2024; 13:e2400581. [PMID: 38637323 DOI: 10.1002/adhm.202400581] [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: 02/15/2024] [Revised: 04/01/2024] [Indexed: 04/20/2024]
Abstract
Macrophages play an essential role in immunotherapy and tissue regeneration owing to their remarkable plasticity and diverse functions. Recent bioengineering developments have focused on using external physical stimuli such as electric and magnetic fields, temperature, and compressive stress, among others, on micro/nanostructures to induce macrophage polarization, thereby increasing their therapeutic potential. However, it is difficult to find a concise review of the interaction between physical stimuli, advanced micro/nanostructures, and macrophage polarization. This review examines the present research on physical stimuli-induced macrophage polarization on micro/nanoplatforms, emphasizing the synergistic role of fabricated structure and stimulation for advanced immunotherapy and tissue regeneration. A concise overview of the research advancements investigating the impact of physical stimuli, including electric fields, magnetic fields, compressive forces, fluid shear stress, photothermal stimuli, and multiple stimulations on the polarization of macrophages within complex engineered structures, is provided. The prospective implications of these strategies in regenerative medicine and immunotherapeutic approaches are highlighted. This review will aid in creating stimuli-responsive platforms for immunomodulation and tissue regeneration.
Collapse
Affiliation(s)
- Keya Ganguly
- Department of Biosystems Engineering, Kangwon National University, Chuncheon, 24341, Republic of Korea
- Institute of Forest Science, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Rachmi Luthfikasari
- Department of Biosystems Engineering, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Aayushi Randhawa
- Department of Biosystems Engineering, Kangwon National University, Chuncheon, 24341, Republic of Korea
- Interdisciplinary Program in Smart Agriculture, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Sayan Deb Dutta
- Department of Biosystems Engineering, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Tejal V Patil
- Department of Biosystems Engineering, Kangwon National University, Chuncheon, 24341, Republic of Korea
- Interdisciplinary Program in Smart Agriculture, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Rumi Acharya
- Department of Biosystems Engineering, Kangwon National University, Chuncheon, 24341, Republic of Korea
- Interdisciplinary Program in Smart Agriculture, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Ki-Taek Lim
- Department of Biosystems Engineering, Kangwon National University, Chuncheon, 24341, Republic of Korea
- Interdisciplinary Program in Smart Agriculture, Kangwon National University, Chuncheon, 24341, Republic of Korea
| |
Collapse
|
38
|
Zeng YY, Gu Q, Li D, Li AX, Liu RM, Liang JY, Liu JY. Immunocyte membrane-derived biomimetic nano-drug delivery system: a pioneering platform for tumour immunotherapy. Acta Pharmacol Sin 2024:10.1038/s41401-024-01355-z. [PMID: 39085407 DOI: 10.1038/s41401-024-01355-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 07/03/2024] [Indexed: 08/02/2024] Open
Abstract
Tumor immunotherapy characterized by its high specificity and minimal side effects has achieved revolutionary progress in the field of cancer treatment. However, the complex mechanisms of tumor immune microenvironment (TIME) and the individual variability of patients' immune system still present significant challenges to its clinical application. Immunocyte membrane-coated nanocarrier systems, as an innovative biomimetic drug delivery platform, exhibit remarkable advantages in tumor immunotherapy due to their high targeting capability, good biocompatibility and low immunogenicity. In this review we summarize the latest research advances in biomimetic delivery systems based on immune cells for tumor immunotherapy. We outline the existing methods of tumor immunotherapy including immune checkpoint therapy, adoptive cell transfer therapy and cancer vaccines etc. with a focus on the application of various immunocyte membranes in tumor immunotherapy and their prospects and challenges in drug delivery and immune modulation. We look forward to further exploring the application of biomimetic delivery systems based on immunocyte membrane-coated nanoparticles, aiming to provide a new framework for the clinical treatment of tumor immunity.
Collapse
Affiliation(s)
- Yuan-Ye Zeng
- School of Pharmacy, Fudan University, Shanghai, 201203, China
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Qing Gu
- Department of Pharmacy, Jingan District Zhabei Central Hospital, Shanghai, 200070, China
| | - Dan Li
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Ai-Xue Li
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Rong-Mei Liu
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Jian-Ying Liang
- School of Pharmacy, Fudan University, Shanghai, 201203, China.
| | - Ji-Yong Liu
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
39
|
Xing Y, Huang Y, Tang Z, Lin Y, Zou Y, Huang Y, He Z, Huang Q, Wu J. Impact of mitochondrial damage on tumor microenvironment and immune response: a comprehensive bibliometric analysis. Front Immunol 2024; 15:1442027. [PMID: 39104527 PMCID: PMC11298338 DOI: 10.3389/fimmu.2024.1442027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Accepted: 07/03/2024] [Indexed: 08/07/2024] Open
Abstract
Background Mitochondrial damage contributes to apoptosis, oxidative stress, and inflammation, which collectively impact the immune system's function and the tumor microenvironment (TME). These processes, in turn, influence tumor cell growth, migration, and response to treatment. Objective We conducted a bibliometric analysis to elucidate the complex interactions between mitochondrial damage, the immune system, and the TME. Methods Data were sourced from the Science Citation Index Core Collection (WoSCC) and analyzed using advanced tools like VOSviewer and Citespace. Our focus was on literature published between 1999 and 2023 concerning the interactions between mitochondrial damage and the TME, as well as immune responses to tumors. The analysis included regional contributions, journal influence, institutional collaborations, authorship, co-cited authors, and keyword citation bursts. Results Our research encompassed 2,039 publications, revealing an increasing trend in annual output exploring the relationship between mitochondrial damage, TME dynamics, and immune responses. China, the United States, and South Korea emerged as the leading contributors. Prominent institutions included Institut National de la Santé et de la Recherche Médicale, University of Texas System, China Medical University, and Sun Yat-sen University. Key journals in this field are the International Journal of Molecular Sciences, Mitochondrion, and the European Journal of Pharmacology. Liang H and Wallace DC were identified as the most productive and co-cited authors, respectively. Keyword analysis highlighted the critical roles of inflammatory responses, oxidative stress, and the immune system in recent research. Conclusion This bibliometric analysis provides a comprehensive overview of historical and current research trends, underscoring the pivotal role of mitochondrial damage in the TME and immune system.
Collapse
Affiliation(s)
- Yichun Xing
- Department of Obstetrics and Gynecology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Yi Huang
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Department of Urology, The Third People’s Hospital of Chengdu/The Affiliated Hospital of Southwest Jiaotong University, Chengdu, Sichuan, China
| | - Zhicheng Tang
- Department of Urology, the Eighth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Ying Lin
- Department of Endocrinology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Yitong Zou
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Yaqiang Huang
- Department of Urology, Zhongshan City People’s Hospital, Sunwen East Road, Zhongshan, Guangdong, China
| | - Zhaohui He
- Department of Urology, the Eighth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Qunxiong Huang
- Department of Urology, the Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Jieying Wu
- Department of Urology, the Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| |
Collapse
|
40
|
Jiang W, Liu Y, Wu Y, Zhang L, Zhang B, Zhou S, Zhang P, Xu T, Wu M, Lv S. Polystyrene nanoplastics of different particle sizes regulate the polarization of pro-inflammatory macrophages. Sci Rep 2024; 14:16329. [PMID: 39009713 PMCID: PMC11251024 DOI: 10.1038/s41598-024-67289-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: 03/13/2024] [Accepted: 07/09/2024] [Indexed: 07/17/2024] Open
Abstract
Microplastics (MPs) are defined as plastic particles smaller than 5 mm in size, and nanoplastics (NPs) are those MPs with a particle size of less than 1000 nm or 100 nm. The prevalence of MPs in the environment and human tissues has raised concerns about their potential negative effects on human health. Macrophages are the major defence against foreign substances in the intestine, and can be polarized into two types: the M1 phenotype and the M2 phenotype. However, the effect of NPs on the polarization of macrophages remains unclear. Herein, we selected polystyrene, one of the most plastics in the environment and controlled the particle sizes at 50 nm and 500 nm respectively to study the effects on the polarization of macrophages. We used mouse RAW264.7 cell line models in this macrophage-associated study. Experiments on cell absorption showed that macrophages could quickly ingest polystyrene nanoplastics of both diameters with time-dependent uptake. Compared to the untreated group and 10 μg/mL treatment group, macrophages exposed to 50 μg/mL groups (50 nm and 500 nm) had considerably higher levels of CD86, iNOS, and TNF-α, but decreased levels of aCD206, IL-10, and Arg-1. According to these findings, macrophage M1 and M2 polarization can both be induced and inhibited by 50 μg/mL 50 nm and 500 nm polystyrene nanoplastics. This work provided the first evidence of a possible MPs mode of action with appropriate concentration and size through the production of polarized M1, providing dietary and environmental recommendations for people, particularly those with autoimmune and autoinflammatory illnesses.
Collapse
Affiliation(s)
- Wanlan Jiang
- Department of Rheumatology and Immunology, The First People's Hospital of Changzhou (The Third Affiliated Hospital of Soochow University), Changzhou, 213003, China
| | - Yilin Liu
- School of Pharmacy, Changzhou University, Changzhou, 213164, China
| | - Yuqi Wu
- School of Medicine, Nantong University, Nantong, 226001, China
| | - Lu Zhang
- Department of Rheumatology and Immunology, The First People's Hospital of Changzhou (The Third Affiliated Hospital of Soochow University), Changzhou, 213003, China
| | - Biqing Zhang
- Department of Rheumatology and Immunology, The First People's Hospital of Changzhou (The Third Affiliated Hospital of Soochow University), Changzhou, 213003, China
| | - Shiliang Zhou
- Department of Rheumatology and Immunology, The First People's Hospital of Changzhou (The Third Affiliated Hospital of Soochow University), Changzhou, 213003, China
| | - Peng Zhang
- School of Materials Science and Engineering, Changzhou University, Changzhou, 213164, China
| | - Ting Xu
- Department of Rheumatology and Immunology, The First People's Hospital of Changzhou (The Third Affiliated Hospital of Soochow University), Changzhou, 213003, China
| | - Min Wu
- Department of Rheumatology and Immunology, The First People's Hospital of Changzhou (The Third Affiliated Hospital of Soochow University), Changzhou, 213003, China.
| | - Songwei Lv
- School of Pharmacy, Changzhou University, Changzhou, 213164, China.
| |
Collapse
|
41
|
Wang FX, Shi ZA, Mu G. Regulation of immune cells by miR-451 and its potential as a biomarker in immune-related disorders: a mini review. Front Immunol 2024; 15:1421473. [PMID: 39076992 PMCID: PMC11284029 DOI: 10.3389/fimmu.2024.1421473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 07/01/2024] [Indexed: 07/31/2024] Open
Abstract
In 2005, Altuvia and colleagues were the first to identify the gene that encodes miR-451 in the human pituitary gland, located in chromosome region 17q11.2. Subsequent studies have confirmed that miR-451 regulates various immune cells, including T cells, B cells, microglia, macrophages, and neutrophils, thereby influencing disease progression. The range of immune-related diseases affected encompasses various cancers, lymphoblastic leukemia, and injuries to the lungs and spinal cord, among others. Moreover, miR-451 is produced by immune cells and can regulate both their own functions and those of other immune cells, thus creating a regulatory feedback loop. This article aims to comprehensively review the interactions between miR-451 and immune cells, clarify the regulatory roles of miR-451 within the immune system, and assess its potential as both a therapeutic target and a biomarker for immune-related diseases.
Collapse
Affiliation(s)
- Fei-xiang Wang
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University, Luzhou, Sichuan, China
| | - Zu-an Shi
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University, Luzhou, Sichuan, China
| | - Guo Mu
- Department of Anesthesiology, Zigong Fourth People’s Hospital, Zigong, Sichuan, China
| |
Collapse
|
42
|
Sun M, Li Y, Xu G, Zhu J, Lu R, An S, Zeng Z, Deng Z, Cheng R, Zhang Q, Yao Y, Wu J, Zhang Y, Hu H, Chen Z, Huang Q, Wu J. Sirt3-Mediated Opa1 Deacetylation Protects Against Sepsis-Induced Acute Lung Injury by Inhibiting Alveolar Macrophage Pro-Inflammatory Polarization. Antioxid Redox Signal 2024. [PMID: 38874521 DOI: 10.1089/ars.2023.0322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/15/2024]
Abstract
Aims: Mitochondrial dynamics in alveolar macrophages (AMs) are associated with sepsis-induced acute lung injury (ALI). In this study, we aimed to investigate whether changes in mitochondrial dynamics could alter the polarization of AMs in sepsis-induced ALI and to explore the regulatory mechanism of mitochondrial dynamics by focusing on sirtuin (SIRT)3-induced optic atrophy protein 1 (OPA1) deacetylation. Results: The AMs of sepsis-induced ALI showed imbalanced mitochondrial dynamics and polarization to the M1 macrophage phenotype. In sepsis, SIRT3 overexpression promotes mitochondrial dynamic equilibrium in AMs. However, 3-(1H-1, 2, 3-triazol-4-yl) pyridine (3TYP)-specific inhibition of SIRT3 increased the mitochondrial dynamic imbalance and pro-inflammatory polarization of AMs and further aggravated sepsis-induced ALI. OPA1 is directly bound to and deacetylated by SIRT3 in AMs. In AMs of sepsis-induced ALI, SIRT3 protein expression was decreased and OPA1 acetylation was increased. OPA1 acetylation at the lysine 792 amino acid residue (OPA1-K792) promotes self-cleavage and is associated with an imbalance in mitochondrial dynamics. However, decreased acetylation of OPA1-K792 reversed the pro-inflammatory polarization of AMs and protected the barrier function of alveolar epithelial cells in sepsis-induced ALI. Innovation: Our study revealed, for the first time, the regulation of mitochondrial dynamics and AM polarization by SIRT3-mediated deacetylation of OPA1 in sepsis-induced ALI, which may serve as an intervention target for precision therapy of the disease. Conclusions: Our data suggest that imbalanced mitochondrial dynamics promote pro-inflammatory polarization of AMs in sepsis-induced ALI and that deacetylation of OPA1 mediated by SIRT3 improves mitochondrial dynamic equilibrium, thereby ameliorating lung injury.
Collapse
Affiliation(s)
- Maomao Sun
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Yuying Li
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Anesthesiology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Gege Xu
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Junrui Zhu
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Ruimin Lu
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Sheng An
- Department of Anesthesiology, The Affiliated Hospital of Guizhou Medical University, Guiyang, People's Republic of China
| | - Zhenhua Zeng
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhiya Deng
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ran Cheng
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Qin Zhang
- Department of Anesthesiology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Yi Yao
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Junjie Wu
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yuan Zhang
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hongbin Hu
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhongqing Chen
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qiaobing Huang
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Jie Wu
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
43
|
Lopes FB, Sarandy MM, Novaes RD, Valacchi G, Gonçalves RV. OxInflammatory Responses in the Wound Healing Process: A Systematic Review. Antioxidants (Basel) 2024; 13:823. [PMID: 39061892 PMCID: PMC11274091 DOI: 10.3390/antiox13070823] [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: 05/03/2024] [Revised: 06/11/2024] [Accepted: 06/27/2024] [Indexed: 07/28/2024] Open
Abstract
Significant sums are spent every year to find effective treatments to control inflammation and speed up the repair of damaged skin. This study investigated the main mechanisms involved in the skin wound cure. Consequently, it offered guidance to develop new therapies to control OxInflammation and infection and decrease functional loss and cost issues. This systematic review was conducted using the PRISMA guidelines, with a structured search in the MEDLINE (PubMed), Scopus, and Web of Science databases, analyzing 23 original studies. Bias analysis and study quality were assessed using the SYRCLE tool (Prospero number is CRD262 936). Our results highlight the activation of membrane receptors (IFN-δ, TNF-α, toll-like) in phagocytes, especially macrophages, during early wound healing. The STAT1, IP3, and NF-kβ pathways are positively regulated, while Ca2+ mobilization correlates with ROS production and NLRP3 inflammasome activation. This pathway activation leads to the proteolytic cleavage of caspase-1, releasing IL-1β and IL-18, which are responsible for immune modulation and vasodilation. Mediators such as IL-1, iNOS, TNF-α, and TGF-β are released, influencing pro- and anti-inflammatory cascades, increasing ROS levels, and inducing the oxidation of lipids, proteins, and DNA. During healing, the respiratory burst depletes antioxidant defenses (SOD, CAT, GST), creating a pro-oxidative environment. The IFN-δ pathway, ROS production, and inflammatory markers establish a positive feedback loop, recruiting more polymorphonuclear cells and reinforcing the positive interaction between oxidative stress and inflammation. This process is crucial because, in the immune system, the vicious positive cycle between ROS, the oxidative environment, and, above all, the activation of the NLRP3 inflammasome inappropriately triggers hypoxia, increases ROS levels, activates pro-inflammatory cytokines and inhibits the antioxidant action and resolution of anti-inflammatory cytokines, contributing to the evolution of chronic inflammation and tissue damage.
Collapse
Affiliation(s)
- Fernanda Barbosa Lopes
- Department of General Biology, Federal University of Viçosa, Viçosa 36570-900, Minas Gerais, Brazil
| | - Mariáurea Matias Sarandy
- Department of General Biology, Federal University of Viçosa, Viçosa 36570-900, Minas Gerais, Brazil
- Plants for Human Health Institute, Animal Science Department, North Carolina State University, North Carolina Research Campus, Kannapolis, NC 28081, USA
| | - Rômulo Dias Novaes
- Department of Structural Biology, Federal University of Alfenas, Alfenas 37130-001, Minas Gerais, Brazil
- Department of Animal Biology, Federal University of Viçosa, Viçosa 36570-900, Minas Gerais, Brazil
| | - Giuseppe Valacchi
- Plants for Human Health Institute, Animal Science Department, North Carolina State University, North Carolina Research Campus, Kannapolis, NC 28081, USA
- Department of Environmental and Prevention Sciences, University of Ferrara, 44121 Ferrara, Italy
- Department of Food and Nutrition, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Reggiani Vilela Gonçalves
- Department of General Biology, Federal University of Viçosa, Viçosa 36570-900, Minas Gerais, Brazil
- Plants for Human Health Institute, Animal Science Department, North Carolina State University, North Carolina Research Campus, Kannapolis, NC 28081, USA
- Department of Animal Biology, Federal University of Viçosa, Viçosa 36570-900, Minas Gerais, Brazil
| |
Collapse
|
44
|
Prapas P, Anagnostouli M. Macrophages and HLA-Class II Alleles in Multiple Sclerosis: Insights in Therapeutic Dynamics. Int J Mol Sci 2024; 25:7354. [PMID: 39000461 PMCID: PMC11242320 DOI: 10.3390/ijms25137354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 07/01/2024] [Accepted: 07/02/2024] [Indexed: 07/16/2024] Open
Abstract
Antigen presentation is a crucial mechanism that drives the T cell-mediated immune response and the development of Multiple Sclerosis (MS). Genetic alterations within the highly variable Major Histocompatibility Complex Class II (MHC II) have been proven to result in significant changes in the molecular basis of antigen presentation and the clinical course of patients with both Adult-Onset MS (AOMS) and Pediatric-Onset MS (POMS). Among the numerous polymorphisms of the Human Leucocyte Antigens (HLA), within MHC II complex, HLA-DRB1*15:01 has been labeled, in Caucasian ethnic groups, as a high-risk allele for MS due to the ability of its structure to increase affinity to Myelin Basic Protein (MBP) epitopes. This characteristic, among others, in the context of the trimolecular complex or immunological synapsis, provides the foundation for autoimmunity triggered by environmental or endogenous factors. As with all professional antigen presenting cells, macrophages are characterized by the expression of MHC II and are often implicated in the formation of MS lesions. Increased presence of M1 macrophages in MS patients has been associated both with progression and onset of the disease, each involving separate but similar mechanisms. In this critical narrative review, we focus on macrophages, discussing how HLA genetic alterations can promote dysregulation of this population's homeostasis in the periphery and the Central Nervous System (CNS). We also explore the potential interconnection in observed pathological macrophage mechanisms and the function of the diverse structure of HLA alleles in neurodegenerative CNS, seen in MS, by comparing available clinical with molecular data through the prism of HLA-immunogenetics. Finally, we discuss available and experimental pharmacological approaches for MS targeting the trimolecular complex that are based on cell phenotype modulation and HLA genotype involvement and try to reveal fertile ground for the potential development of novel drugs.
Collapse
Affiliation(s)
- Petros Prapas
- Research Immunogenetics Laboratory, First Department of Neurology, Aeginition University Hospital, School of Medicine, National and Kapodistrian University of Athens, Vas. Sofias 72-74, 11528 Athens, Greece
| | - Maria Anagnostouli
- Research Immunogenetics Laboratory, First Department of Neurology, Aeginition University Hospital, School of Medicine, National and Kapodistrian University of Athens, Vas. Sofias 72-74, 11528 Athens, Greece
- Multiple Sclerosis and Demyelinating Diseases Unit, Center of Expertise for Rare Demyelinating and Autoimmune Diseases of CNS, First Department of Neurology, School of Medicine, National and Kapodistrian University of Athens NKUA, Aeginition University Hospital, Vas. Sofias 72-74, 11528 Athens, Greece
| |
Collapse
|
45
|
Binder AK, Bremm F, Dörrie J, Schaft N. Non-Coding RNA in Tumor Cells and Tumor-Associated Myeloid Cells-Function and Therapeutic Potential. Int J Mol Sci 2024; 25:7275. [PMID: 39000381 PMCID: PMC11242727 DOI: 10.3390/ijms25137275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 06/19/2024] [Accepted: 06/29/2024] [Indexed: 07/16/2024] Open
Abstract
The RNA world is wide, and besides mRNA, there is a variety of other RNA types, such as non-coding (nc)RNAs, which harbor various intracellular regulatory functions. This review focuses on small interfering (si)RNA and micro (mi)RNA, which form a complex network regulating mRNA translation and, consequently, gene expression. In fact, these RNAs are critically involved in the function and phenotype of all cells in the human body, including malignant cells. In cancer, the two main targets for therapy are dysregulated cancer cells and dysfunctional immune cells. To exploit the potential of mi- or siRNA therapeutics in cancer therapy, a profound understanding of the regulatory mechanisms of RNAs and following targeted intervention is needed to re-program cancer cells and immune cell functions in vivo. The first part focuses on the function of less well-known RNAs, including siRNA and miRNA, and presents RNA-based technologies. In the second part, the therapeutic potential of these technologies in treating cancer is discussed, with particular attention on manipulating tumor-associated immune cells, especially tumor-associated myeloid cells.
Collapse
Affiliation(s)
- Amanda Katharina Binder
- Department of Dermatology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany; (A.K.B.); (F.B.); (J.D.)
- Comprehensive Cancer Center Erlangen European Metropolitan Area of Nuremberg (CCC ER-EMN), 91054 Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), 91054 Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), 91054 Erlangen, Germany
| | - Franziska Bremm
- Department of Dermatology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany; (A.K.B.); (F.B.); (J.D.)
- Comprehensive Cancer Center Erlangen European Metropolitan Area of Nuremberg (CCC ER-EMN), 91054 Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), 91054 Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), 91054 Erlangen, Germany
| | - Jan Dörrie
- Department of Dermatology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany; (A.K.B.); (F.B.); (J.D.)
- Comprehensive Cancer Center Erlangen European Metropolitan Area of Nuremberg (CCC ER-EMN), 91054 Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), 91054 Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), 91054 Erlangen, Germany
| | - Niels Schaft
- Department of Dermatology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany; (A.K.B.); (F.B.); (J.D.)
- Comprehensive Cancer Center Erlangen European Metropolitan Area of Nuremberg (CCC ER-EMN), 91054 Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), 91054 Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), 91054 Erlangen, Germany
| |
Collapse
|
46
|
Song CY, Wu CY, Lin CY, Tsai CH, Chen HT, Fong YC, Chen LC, Tang CH. The stimulation of exosome generation by visfatin polarizes M2 macrophages and enhances the motility of chondrosarcoma. ENVIRONMENTAL TOXICOLOGY 2024; 39:3790-3798. [PMID: 38497692 DOI: 10.1002/tox.24236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 02/25/2024] [Accepted: 03/07/2024] [Indexed: 03/19/2024]
Abstract
Chondrosarcoma is a malignant bone tumor that arises from abnormalities in cartilaginous tissue and is associated with lung metastases. Extracellular vesicles called exosomes are primarily used as mediators of intercellular signal transmission to control tumor metastasis. Visfatin is an adipokine reported to enhance tumor metastasis, but its relationship with exosome generation in chondrosarcoma motility remains undetermined. Our results found that overexpressing visfatin augments the production of exosomes from chondrosarcoma cells. Visfatin-treated chondrosarcoma exosomes educate macrophage polarization towards the M2 but not M1 phenotype. Interestingly, M2 macrophages polarized by exosomes return to chondrosarcoma cells to facilitate cell motility. According to these findings, chondrosarcoma cells emit more exosomes when treated with visfatin. The stimulation of exosome generation by visfatin polarizes M2 macrophages and enhances the motility of chondrosarcoma.
Collapse
Affiliation(s)
- Chang-Yu Song
- Graduate Institute of Biomedical Science, China Medical University, Taichung, Taiwan
| | - Chih-Ying Wu
- Graduate Institute of Integrated Medicine, China Medical University, Taichung, Taiwan
- Department of Neurosurgery, China Medical University Hospital, Taichung, Taiwan
- Department of Neurosurgery, China Medical University Hsinchu Hospital, Hsinchu, Taiwan
| | - Chih-Yang Lin
- Translational Medicine Center, Shin-Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
| | - Chun-Hao Tsai
- Department of Sports Medicine, College of Health Care, China Medical University, Taichung, Taiwan
- Department of Orthopedic Surgery, China Medical University Hospital, Taichung, Taiwan
| | - Hsien-Te Chen
- Department of Sports Medicine, College of Health Care, China Medical University, Taichung, Taiwan
- Department of Orthopedic Surgery, China Medical University Hospital, Taichung, Taiwan
| | - Yi-Chin Fong
- Department of Sports Medicine, College of Health Care, China Medical University, Taichung, Taiwan
- Department of Orthopedic Surgery, China Medical University Hospital, Taichung, Taiwan
- Department of Orthopedic Surgery, China Medical University Beigang Hospital, Taichung, Yunlin, Taiwan
| | - Li-Chai Chen
- Department of Pharmacy, Tajen University, Pingtung, Taiwan
| | - Chih-Hsin Tang
- Graduate Institute of Biomedical Science, China Medical University, Taichung, Taiwan
- Department of Pharmacology, School of Medicine, China Medical University, Taichung, Taiwan
- Department of Medical Laboratory Science and Biotechnology, College of Medical and Health Science, Asia University, Taichung, Taiwan
- Chinese Medicine Research Center, China Medical University, Taichung, Taiwan
- Department of Medical Research, China Medical University Hsinchu Hospital, Hsinchu, Taiwan
| |
Collapse
|
47
|
Zhang B, Wang W, Gao P, Li X, Chen L, Lin Z, Chen H, Liang W, Kong Z, Lin D, Wu X, Zhang T. Injectable, Electroconductive, Free Radical Scavenging Silk Fibroin/Black Phosphorus/Glycyrrhizic Acid Nanocomposite Hydrogel for Enhancing Spinal Cord Repair. Adv Healthc Mater 2024; 13:e2304300. [PMID: 38589053 DOI: 10.1002/adhm.202304300] [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: 12/04/2023] [Revised: 03/25/2024] [Indexed: 04/10/2024]
Abstract
Spinal cord injury (SCI) often leads to a severe permanent disability. A poor inflammatory microenvironment and nerve electric signal conduction block are the main reasons for difficulty in spinal cord nerve regeneration. In this study, black phosphorus (BP) and glycyrrhizic acid (GA) are integrated into methacrylate-modified silk fibroin (SF) to construct a bifunctional injectable hydrogel (SF/BP/GA) with appropriate conductivity and the ability to inhibit inflammation to promote neuronal regeneration after SCI. This work discovers that the SF/BP/GA hydrogel can reduce the oxidative damage mediated by oxygen free radicals, promote the polarization of macrophages toward the anti-inflammatory M2 phenotype, reduce the expression of inflammatory factors, and improve the inflammatory microenvironment. Moreover, it induces neural stem cell (NSC) differentiation and neurosphere formation, restores signal conduction at the SCI site in vivo, and ameliorates motor function in mice with spinal cord hemisection, revealing a significant neural repair effect. An injectable, electroconductive, free-radical-scavenging hydrogel is a promising therapeutic strategy for SCI repair.
Collapse
Affiliation(s)
- Beichen Zhang
- Department of Graduate School and Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- Department of Orthopedic Surgery, Department of Neurosurgery, General Hospital of Southern Theatre Command of PLA, Guangzhou, 510010, China
| | - Wanshun Wang
- Department of Graduate School and Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- Department of Orthopedic Surgery, Department of Neurosurgery, General Hospital of Southern Theatre Command of PLA, Guangzhou, 510010, China
- Department of Orthopedic Surgery, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, 510120, China
| | - Peng Gao
- Department of Graduate School and Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- Department of Orthopedic Surgery, Department of Neurosurgery, General Hospital of Southern Theatre Command of PLA, Guangzhou, 510010, China
| | - Xiang Li
- Department of Graduate School and Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Lingling Chen
- Department of Orthopedic Surgery, Department of Neurosurgery, General Hospital of Southern Theatre Command of PLA, Guangzhou, 510010, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Zefeng Lin
- Department of Orthopedic Surgery, Department of Neurosurgery, General Hospital of Southern Theatre Command of PLA, Guangzhou, 510010, China
| | - Hu Chen
- Department of Orthopedic Surgery, Department of Neurosurgery, General Hospital of Southern Theatre Command of PLA, Guangzhou, 510010, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Wenhao Liang
- Department of Orthopedic Surgery, Department of Neurosurgery, General Hospital of Southern Theatre Command of PLA, Guangzhou, 510010, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Zhiyuan Kong
- Department of Orthopedic Surgery, Department of Neurosurgery, General Hospital of Southern Theatre Command of PLA, Guangzhou, 510010, China
| | - Dingkun Lin
- Department of Graduate School and Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- Department of Orthopedic Surgery, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, 510120, China
| | - Xiaona Wu
- Department of Orthopedic Surgery, Department of Neurosurgery, General Hospital of Southern Theatre Command of PLA, Guangzhou, 510010, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Tao Zhang
- Department of Graduate School and Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- Department of Orthopedic Surgery, Department of Neurosurgery, General Hospital of Southern Theatre Command of PLA, Guangzhou, 510010, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| |
Collapse
|
48
|
Dorry S, Perla S, Bennett AM. MAPK Phosphatase-5 is required for TGF-β signaling through a JNK-dependent pathway. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.27.600976. [PMID: 38979264 PMCID: PMC11230413 DOI: 10.1101/2024.06.27.600976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Mitogen-activated protein kinase (MAPK) phosphatases (MKPs) constitute members of the dual-specificity family of protein phosphatases that dephosphorylate the MAPKs. MKP-5 dephosphorylates the stress-responsive MAPKs, p38 MAPK and JNK, and has been shown to promote tissue fibrosis. Here, we provide insight into how MKP-5 regulates the transforming growth factor-β (TGF-β) pathway, a well-established driver of fibrosis. We show that MKP-5-deficient fibroblasts in response to TGF-β are impaired in SMAD2 phosphorylation at canonical and non-canonical sites, nuclear translocation, and transcriptional activation of fibrogenic genes. Consistent with this, pharmacological inhibition of MKP-5 is sufficient to block TGF-β signaling, and that this regulation occurs through a JNK-dependent pathway. By utilizing RNA sequencing and transcriptomic analysis, we identify TGF-β signaling activators regulated by MKP-5 in a JNK-dependent manner, providing mechanistic insight into how MKP-5 promotes TGF-β signaling. This study elucidates a novel mechanism whereby MKP-5-mediated JNK inactivation is required for TGF-β signaling and provides insight into the role of MKP-5 in fibrosis.
Collapse
Affiliation(s)
- Sam Dorry
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Sravan Perla
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Anton M. Bennett
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut, USA
- Yale Center for Molecular and Systems Metabolism, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
49
|
Fang T, Chen G. Non-viral vector-based genome editing for cancer immunotherapy. Biomater Sci 2024; 12:3068-3085. [PMID: 38716572 DOI: 10.1039/d4bm00286e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2024]
Abstract
Despite the exciting promise of cancer immunotherapy in the clinic, immune checkpoint blockade therapy and T cell-based therapies are often associated with low response rates, intrinsic and adaptive immune resistance, and systemic side effects. CRISPR-Cas-based genome editing appears to be an effective strategy to overcome these unmet clinical needs. As a safer delivery platform for the CRISPR-Cas system, non-viral nanoformulations have been recently explored to target tumor cells and immune cells, aiming to improve cancer immunotherapy on a gene level. In this review, we summarized the efforts of non-viral vector-based CRISPR-Cas-mediated genome editing in tumor cells and immune cells for cancer immunotherapy. Their design rationale and specific applications were highlighted.
Collapse
Affiliation(s)
- Tianxu Fang
- Department of Biomedical Engineering, McGill University, Montreal, QC, H3G 0B1, Canada.
- Rosalind & Morris Goodman Cancer Institute, McGill University, Montreal, QC, H3G 0B1, Canada
| | - Guojun Chen
- Department of Biomedical Engineering, McGill University, Montreal, QC, H3G 0B1, Canada.
- Rosalind & Morris Goodman Cancer Institute, McGill University, Montreal, QC, H3G 0B1, Canada
| |
Collapse
|
50
|
Wang W, Wan F, Yu T, Wu S, Cui X, Xiang C, Li M, Liu Q, Lin C. Microvesicles-delivering Smad7 have advantages over microvesicles in suppressing fibroblast differentiation in a model of Peyronie's disease. BMC Biotechnol 2024; 24:40. [PMID: 38849776 PMCID: PMC11162046 DOI: 10.1186/s12896-024-00866-1] [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: 04/08/2024] [Accepted: 06/04/2024] [Indexed: 06/09/2024] Open
Abstract
BACKGROUND This study compared the differences of microvesicles (MVs) and microvesicles-delivering Smad7 (Smad7-MVs) on macrophage M1 polarization and fibroblast differentiation in a model of Peyronie's disease (PD). METHODS Overexpression of Smad7 in rat BMSCs was obtained by pCMV5-Smad7 transfection. MVs were collected from rat BMSCs using ultracentrifugation. In cells, 100 µg/mL of MVs or Smad7-MVs were used to treat the 100 ng/mL of lipopolysaccharide (LPS)-induced RAW264.7 cells or 10 ng/mL of recombinant transforming growth factor-β1 (TGF-β1)-induced fibroblasts. The pro-inflammatory cytokines and markers of M1 macrophages were measured in RAW264.7 cells, and the migration and markers of fibroblast differentiation were measured in fibroblasts. In rats, 50 µg of MVs or Smad7-MVs were used to treat the TGF-β1-induced animals. The pathology of tunica albuginea (TA), the markers of M1 macrophages and fibroblast differentiation in the TA were measured. RESULTS The MVs or Smad7-MVs treatment suppressed the LPS-induced macrophage M1 polarization and TGF-β1-induced fibroblast differentiation. Moreover, the Smad7-MVs treatment decreased the fibroblast differentiation compared with the MVs treatment. In the TGF-β1-induced TA of rats, MVs or Smad7-MVs treatment ameliorated the TA fibrosis by suppressing the macrophage M1 polarization and fibroblast differentiation. There was no significance on the M1-polarized macrophages between the MVs treatment and the Smad7-MVs treatment. Meanwhile, the Smad7-MVs treatment had an edge in terms of suppressing the fibroblast differentiation in the TGF-β1-induced PD model compared with the MVs treatment. CONCLUSIONS This study demonstrated that Smad7-MVs treatment had advantages over MVs treatment in suppressing of fibroblast differentiation in a model of PD.
Collapse
Affiliation(s)
- Wenting Wang
- Central Laboratory, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, 264000, China
| | - Fengchun Wan
- Organ Transplant Center, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, 264000, China
| | - Tianxi Yu
- School of Clinical Medicine, Weifang Medical University, Weifang, 261042, China
- Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, 264000, China
| | - Shuang Wu
- Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, 264000, China
| | - Xin Cui
- School of Clinical Medicine, Weifang Medical University, Weifang, 261042, China
- Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, 264000, China
| | - Chongjun Xiang
- Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, 264000, China
- The 2nd Medical College of Binzhou Medical University, Yantai, 264003, China
| | - Monong Li
- Department of Urology, Qingdao Municipal Hospital, Qingdao, 266011, China
| | - Qingzuo Liu
- Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, 264000, China.
| | - Chunhua Lin
- Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, 264000, China.
| |
Collapse
|