1
|
Zheng H, Dong Y, Zhang X, Liu J, Geng X, Liu Z, Liao Y, Liu Y, Yang P, Yang G, Liu X. Modulation of Mettl5 alleviates airway allergy by regulating the epigenetic profile of M2 macrophages. Cell Signal 2025; 131:111740. [PMID: 40089091 DOI: 10.1016/j.cellsig.2025.111740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 02/20/2025] [Accepted: 03/10/2025] [Indexed: 03/17/2025]
Abstract
M2 macrophages (M2 cells) are known to be involved in both Th2 responses and immune regulation. However, the underlying mechanisms remain unclear. Functional abnormalities in macrophages are associated with airway allergy (AA). The objective of this study was to investigate the role of methyltransferase-like 5 (Mettl5) in macrophages and its potential to alleviate AA. In this study, an airway allergy (AA) mouse model was established using dust mite extracts (DME) as the specific antigen. M2 cells were collected from mice with and without AA. The role of Mettl5 in modulating the immune activities of M2 cells was assessed using both epigenetic and immunological approaches. We found that Mettl5 levels were elevated in airway M2 cells from mice with AA. The presence of Mettl5 in airway M2 cells was positively correlated with airway Th2 polarization in these mice. Airway M2 cells from AA mice exhibited impaired immune-suppressive function, which was resolved by ablating the Mettl5 gene in macrophages. Mettl5 was responsible for the hypermethylation of the Il10 promoter in airway M2 cells of AA mice. Exposure to DME induced Mettl5, which in turn recruited USP21 to deubiquitinate GATA3, thereby boosting IL-4 expression in M2 cells. Inhibiting Mettl5 restored the immune-suppressive capacity of airway M2 cells and mitigated experimental AA. In conclusion, Mettl5 plays a critical role in subverting the immune-regulatory capacity and enhancing IL-4 expression in M2 cells. Inhibition of Mettl5 can mitigate experimental AA by restoring the immune-regulatory functions of M2 cells.
Collapse
Affiliation(s)
- Haoyue Zheng
- Department of Otolaryngology, Longgang Central Hospital and Clinical College affiliated to Guangzhou University of Chinese Medicine, Shenzhen, China; State Key Laboratory of Respiratory Diseases Allergy Division at Shenzhen University, Institute of Allergy & Immunology, Shenzhen University School of Medicine, and Shenzhen Key Laboratory of Allergy & Immunology, Shenzhen, China
| | - Yixuan Dong
- Department of Otolaryngology, Longgang Central Hospital and Clinical College affiliated to Guangzhou University of Chinese Medicine, Shenzhen, China; State Key Laboratory of Respiratory Diseases Allergy Division at Shenzhen University, Institute of Allergy & Immunology, Shenzhen University School of Medicine, and Shenzhen Key Laboratory of Allergy & Immunology, Shenzhen, China
| | - Xiwen Zhang
- Department of Otolaryngology, Longgang Central Hospital and Clinical College affiliated to Guangzhou University of Chinese Medicine, Shenzhen, China; State Key Laboratory of Respiratory Diseases Allergy Division at Shenzhen University, Institute of Allergy & Immunology, Shenzhen University School of Medicine, and Shenzhen Key Laboratory of Allergy & Immunology, Shenzhen, China
| | - Jiangqi Liu
- Department of Otolaryngology, Longgang ENT Hospital & Shenzhen ENT Institute, Shenzhen, China
| | - Xiaorui Geng
- Department of Otolaryngology, Longgang ENT Hospital & Shenzhen ENT Institute, Shenzhen, China
| | - Zhiqiang Liu
- Department of Otolaryngology, Longgang ENT Hospital & Shenzhen ENT Institute, Shenzhen, China
| | - Yun Liao
- Department of General Practice Medicine, Third Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Yu Liu
- Department of General Practice Medicine, Third Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Pingchang Yang
- State Key Laboratory of Respiratory Diseases Allergy Division at Shenzhen University, Institute of Allergy & Immunology, Shenzhen University School of Medicine, and Shenzhen Key Laboratory of Allergy & Immunology, Shenzhen, China.
| | - Gui Yang
- Department of Otolaryngology, Longgang Central Hospital and Clinical College affiliated to Guangzhou University of Chinese Medicine, Shenzhen, China.
| | - Xiaoyu Liu
- State Key Laboratory of Respiratory Diseases Allergy Division at Shenzhen University, Institute of Allergy & Immunology, Shenzhen University School of Medicine, and Shenzhen Key Laboratory of Allergy & Immunology, Shenzhen, China.
| |
Collapse
|
2
|
Lu J, Zhou Y, Song YX, Wang JY, Xian JX. Natural alkaloids modulating macrophage polarization: Innovative therapeutic strategies for inflammatory, cardiovascular, and cancerous diseases. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156709. [PMID: 40250001 DOI: 10.1016/j.phymed.2025.156709] [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: 12/31/2024] [Revised: 03/21/2025] [Accepted: 03/29/2025] [Indexed: 04/20/2025]
Abstract
BACKGROUND Macrophage polarization, switching between pro-inflammatory M1 and anti-inflammatory M2 states, is crucial for disease dynamics in inflammatory, metabolic, and cancer contexts. Modulating this polarization is a clinical challenge, but natural alkaloids, with their potent anti-inflammatory and immunomodulatory effects, show promise in reprogramming macrophage phenotypes. PURPOSE This review explores the applications of natural alkaloids-such as matrine, berberine, koumine, sophoridine, and curcumin-in modulating macrophage polarization. It aims to highlight their potential in reprogramming macrophage phenotypes and improving therapeutic outcomes across various diseases. METHODS A comprehensive literature review was conducted using databases like PubMed, Web of Science, Science Direct and Google Scholar, employing targeted keywords related to natural alkaloids, macrophage polarization, and disease treatment. The analysis primarily focused on articles published between 2020 and 2024. RESULTS This review summarizes how natural alkaloids regulate macrophage polarization, promoting the M2 phenotype to reduce inflammation, thereby playing a therapeutic role in anti-inflammatory, cardiovascular, and metabolic diseases. At the same time, they also promote M1 polarization to inhibit tumor development. CONCLUSION Accumulating evidence demonstrates that macrophage polarization regulation by natural alkaloids holds notable clinical value for disease intervention. They alleviate inflammation, enhance antitumor immunity, and improve treatment outcomes, demonstrating their importance in innovative therapeutic strategies. Moreover, combining alkaloids with immunotherapy enhances treatment efficacy, further highlighting their versatility in a variety of therapeutic applications.
Collapse
Affiliation(s)
- Jing Lu
- Department of Pharmacy, The Third People's Hospital of Chengdu, Chengdu, Sichuan, China
| | - Ying Zhou
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yi-Xuan Song
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Jie-Ying Wang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Jia-Xun Xian
- Traditional Chinese Medicine Hospital of Meishan, Meishan 620010, China.
| |
Collapse
|
3
|
Zong J, Wang C, Zhou H, Song Y, Fang K, Chang X. ApoE [-/-] CA1-overexpressing knock-in mice aggravated atherosclerosis by increasing M1 macrophages. ATHEROSCLEROSIS PLUS 2025; 60:6-19. [PMID: 40236987 PMCID: PMC11999080 DOI: 10.1016/j.athplu.2025.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 03/02/2025] [Accepted: 03/24/2025] [Indexed: 04/17/2025]
Abstract
Background Carbonic anhydrase I (CA1) has been reported to be a diagnostic and therapeutic target for atherosclerosis (AS). This study aimed to verify the essential role of CA1 in AS progression in CA1-overexpressing mice. Methods A ApoE [-/-] CA1-overexpressing knock-in mouse model was constructed via CRISPR/Cas9-mediated genome engineering. AS was then induced in these transgenic mice via the administration of a high-fat diet, and a second group simultaneously received treatment with methazolamide (MTZ), a carbonic anhydrase inhibitor. Results Compared with ApoE [-/-] mice without CA1 overexpression, CA1-overexpressing mice had a greater average body weight, regardless of whether their treatment with MTZ or their AS induction status. Sudan IV, hematoxylin and eosin and Oil Red O staining revealed more plaques and fat deposits in the cardiac aortas of CA1-overexpressing mice than in those of ordinary ApoE-/- mice when AS was induced. Moreover, the atherogenic index; low-density lipoprotein, total cholesterol and triglyceride levels were significantly elevated, and high-density lipoprotein levels were declined in the peripheral blood of CA1-overexpressing mice than in that of ordinary ApoE [-/-] mice, regardless of whether these animals were induced to AS. Immunohistochemistry, Von Kossa staining and fluorescence immunohistochemistry revealed increases in CA1 expression, calcium deposition and M1 macrophages in the aortic tissues of CA1-overexpressing mice with AS. MTZ treatment significantly suppressed AS pathologies in the above experiments. Conclusion These findings revealed aggravated AS in ApoE [-/-] CA1-overexpressing mice and suggest that CA1 aggravates AS by increasing M1-type macrophages, a proinflammatory macrophage subtype.
Collapse
Affiliation(s)
- Jinbao Zong
- Clinical Laboratory and Qingdao Key Laboratory of Immunodiagnosis, Qingdao Hiser Hospital Affiliated of Qingdao University (Qingdao Traditional Chinese Medicine Hospital), Renmin Road 4, Qingdao, 266000, PR China
| | - Changyuan Wang
- Department of Dermatology, Qingdao Hospital, University of Health and Rehabilitation Sciences(Qingdao Municipal Hospital), Dengyun Road 369, Qingdao, 266000, PR China
| | - Hongji Zhou
- Medical Research Center, The Affiliated Hospital of Qingdao University, Wutaishan Road 1677, Qingdao, 266000, PR China
- Department of Cardiology, Fushun Municipal Central Hospital, Xincheng Road 5, Fushun, Liaoning, 113006, PR China
| | - Yu Song
- Medical Research Center, The Affiliated Hospital of Qingdao University, Wutaishan Road 1677, Qingdao, 266000, PR China
| | - Kehua Fang
- Clinical Laboratory, The Affiliated Hospital of Qingdao University, Wutaishan Road 1677, Qingdao, Shandong, 266000, PR China
| | - Xiaotian Chang
- Medical Research Center, The Affiliated Hospital of Qingdao University, Wutaishan Road 1677, Qingdao, 266000, PR China
| |
Collapse
|
4
|
Goleij P, Rezaee A, Lam HY, Tabari MAK, Ouf N, Alijanzadeh D, Sanaye PM, Larsen DS, Daglia M, Khan H, Sethi G, Kumar AP. From bench to bedside: exploring curcumin-driven signaling pathways in immune cells for cancer management. Inflammopharmacology 2025:10.1007/s10787-025-01739-5. [PMID: 40244492 DOI: 10.1007/s10787-025-01739-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 01/02/2025] [Indexed: 04/18/2025]
Abstract
The use of natural compounds as effective therapeutic agents is an expanding area of health and disease research. Curcumin, a bioactive component derived from the rhizome of the turmeric plant (Curcuma longa L.), has been primarily used in culinary applications for several centuries, but now its potential health benefits are the focus of growing scientific research. Interestingly, some studies have found that curcumin has antitumorigenic effects due to its ability to influence the tumor microenvironment and possibly promote immune system response by modulating specific signaling pathways in immune cells. The interaction of curcumin with immune cells in the field of cancer chemoprevention is a complex area of research. It has been suggested that curcumin might promote T cell recruitment, reduce neutrophil and macrophage accumulation in the tumor microenvironment, and prevent the conversion of infiltrating lymphocytes into immunosuppressive subpopulations. Thus, its possible mechanisms of action also include a shift of the immune balance toward activation by reversing the prevalence of immunosuppressive cells. With innovations and improvements in our understanding of the potential benefits of curcumin on immune cells in cancer prevention and treatment, it is important to have an overview of current findings. Therefore, in this study, we aim to provide a review of the latest discoveries regarding curcumin in the field of cancer and immune cell research.
Collapse
Affiliation(s)
- Pouya Goleij
- Immunology Board for Transplantation and Cell-Based Therapeutics (ImmunoTACT), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
- USERN Office, Kermanshah University of Medical Sciences, Kermanshah, Iran.
- PhytoPharmacology Interest Group (PPIG), Universal Scientific Education and Research, Network (USERN), Tehran, Iran.
| | - Aryan Rezaee
- School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Hiu Yan Lam
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Mohammad Amin Khazeei Tabari
- PhytoPharmacology Interest Group (PPIG), Universal Scientific Education and Research, Network (USERN), Tehran, Iran
- Student Research Committee, School of Medicine, Mazandaran University of Medical Sciences, Mazandaran, Iran
| | - Noureldeen Ouf
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Dorsa Alijanzadeh
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Pantea Majma Sanaye
- PhytoPharmacology Interest Group (PPIG), Universal Scientific Education and Research, Network (USERN), Tehran, Iran
- School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Danaé S Larsen
- School of Chemical Sciences, The University of Auckland, 23 Symonds Street, Auckland, 1010, New Zealand
| | - Maria Daglia
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano 49, 80131, Naples, Italy
- International Research Center for Food Nutrition and Safety, Jiangsu University, Zhenjiang, 212013, China
| | - Haroon Khan
- Department of Pharmacy, Faculty of Chemical and Life Sciences, Abdul Wali Khan University Mardan, Mardan, 23200, Pakistan
- Department of Pharmacy, Korea University, Sejong, 20019, South Korea
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
5
|
Qiu Z, Li Z, Zhang C, Zhao Q, Liu Z, Cheng Q, Zhang J, Lin A, Luo P. NK Cell Senescence in Cancer: From Molecular Mechanisms to Therapeutic Opportunities. Aging Dis 2025:AD.2025.0053. [PMID: 40249925 DOI: 10.14336/ad.2025.0053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2025] [Accepted: 03/13/2025] [Indexed: 04/20/2025] Open
Abstract
P Natural killer (NK) cells function as crucial effectors in the innate immune response against tumors. Nevertheless, NK cell senescence, characterized by phenotypic and functional changes, substantially compromises their antitumor immune response. This review provides a comprehensive summary of the molecular mechanisms governing NK cell senescence and its implications for cancer immunotherapy. We propose a refined definition of NK cell senescence based on distinct biomarkers, including elevated CD57 expression, reduced cytotoxicity, and altered cytokine secretion. Moreover, we investigate the complex interactions between the tumor microenvironment (TME) and NK cell senescence, highlighting the influence of chronic inflammation, immunosuppressive cytokines, and persistent tumor antigenic stimulation. Additionally, this review underscores the potential utility of senescent NK cells as biomarkers for assessing antitumor efficacy and examines the adverse effects of NK cell senescence on cancer immunotherapy. Lastly, we summarize current approaches to mitigate NK cell senescence, such as gene editing techniques and cytokine modulation, which may enhance the efficacy of NK cell-based immunotherapies. By establishing a comprehensive framework for understanding NK cell senescence within the TME, this review aims to guide future research and the development of innovative therapeutic strategies targeting senescent NK cells to improve cancer immunotherapy outcomes.
Collapse
Affiliation(s)
- Zilin Qiu
- Donghai County People's Hospital - Jiangnan University Smart Healthcare Joint Laboratory, Donghai County People's Hospital (Affiliated Kangda College of Nanjing Medical University), Lianyungang, 222000, China
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, Guangdong, China
| | - Zhengrui Li
- Department of Oral and Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, Shanghai, 200011, China
| | - Cangang Zhang
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Qun Zhao
- The Third Department of Surgery, the Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, China
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang 050011, China
- Big data analysis and mining application for precise diagnosis and treatment of gastric cancer Hebei Provincial Engineering Research Center, Shijiazhuang 050011, China
| | - Zaoqu Liu
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Quan Cheng
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Hunan, China
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Jian Zhang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, Guangdong, China
| | - Anqi Lin
- Donghai County People's Hospital - Jiangnan University Smart Healthcare Joint Laboratory, Donghai County People's Hospital (Affiliated Kangda College of Nanjing Medical University), Lianyungang, 222000, China
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, Guangdong, China
| | - Peng Luo
- Donghai County People's Hospital - Jiangnan University Smart Healthcare Joint Laboratory, Donghai County People's Hospital (Affiliated Kangda College of Nanjing Medical University), Lianyungang, 222000, China
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, Guangdong, China
| |
Collapse
|
6
|
Lin A, Ding Y, Li Z, Jiang A, Liu Z, Wong HZH, Cheng Q, Zhang J, Luo P. Glucagon-like peptide 1 receptor agonists and cancer risk: advancing precision medicine through mechanistic understanding and clinical evidence. Biomark Res 2025; 13:50. [PMID: 40140925 PMCID: PMC11948983 DOI: 10.1186/s40364-025-00765-3] [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: 11/29/2024] [Accepted: 03/13/2025] [Indexed: 03/28/2025] Open
Abstract
Glucagon-like peptide-1 receptor agonists (GLP-1RAs) have emerged as a primary first-line treatment for type 2 diabetes. This has raised concerns about their impact on cancer risk, spurring extensive research. This review systematically examines the varied effects of GLP-1RAs on the risk of different types of tumors, including overall cancer risk and specific cancers such as thyroid, pancreatic, reproductive system, liver, and colorectal cancers. The potential biological mechanisms underlying their influence on cancer risk are complex, involving metabolic regulation, direct antitumor effects, immune modulation, and epigenetic changes. A systematic comparison with other antidiabetic agents reveals notable differences in their influence on cancer risk across drug classes. Additionally, critical factors that shape the relationship between GLP-1RAs and cancer risk are thoroughly analyzed, including patient demographics, comorbidities, treatment regimens, and lifestyle factors, offering essential insights for developing individualized treatment protocols. Despite significant research progress, critical gaps remain. Future research should prioritize elucidating the molecular mechanisms behind the antitumor effects, refining individualized treatment strategies, investigating early tumor prevention applications, assessing potential benefits for non-diabetic populations, advancing the development of novel therapies, establishing robust safety monitoring frameworks, and building precision medicine decision-making platforms. These efforts aim to establish novel roles for GLP-1RAs in cancer prevention. and treatment, thereby advancing the progress of precision medicine.
Collapse
Affiliation(s)
- Anqi Lin
- Donghai County People's Hospital - Jiangnan University Smart Healthcare Joint Laboratory, Donghai County People's Hospital (Affiliated Kangda College of Nanjing Medical University), Lianyungang, Jiangsu Province, 222000, China
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, Guangdong, China
| | - Yanxi Ding
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, Guangdong, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Zhengrui Li
- Department of Oral and Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, Shanghai, China
| | - Aimin Jiang
- Department of Urology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Zaoqu Liu
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Hank Z H Wong
- Li Ka Shing, Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Hunan, China.
| | - Jian Zhang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, Guangdong, China.
| | - Peng Luo
- Donghai County People's Hospital - Jiangnan University Smart Healthcare Joint Laboratory, Donghai County People's Hospital (Affiliated Kangda College of Nanjing Medical University), Lianyungang, Jiangsu Province, 222000, China.
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, Guangdong, China.
| |
Collapse
|
7
|
Lyu X, Shi J, Liu Q, Jiang M, Liu X, Li Y, Ding S, Dai X. Immunosuppression of spleen in mice treated with erythropoietin: transcriptomic and immunological analysis. Front Immunol 2025; 16:1560589. [PMID: 40191193 PMCID: PMC11968373 DOI: 10.3389/fimmu.2025.1560589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 03/07/2025] [Indexed: 04/09/2025] Open
Abstract
Background and aim Long term high-dose erythropoietin (EPO) had been reported inducing the formation of abdominal aortic aneurysm (AAA) in mice. When using this model, we found that EPO treated mice showed significant splenomegaly. This is an interesting phenomenon, and its mechanism has not been reported. Therefore, this study aims to explore its mechanism. Methods C57BL/6 mice were given intraperitoneal injection of recombinant human EPO at 10000 IU/kg/day, and the control mice were treated with normal saline (vehicle). After 3 weeks, the spleens were harvested. Pathological changes in histology were observed using Hematoxylin and Eosin (H&E) staining. The differential expression genes (DEGs) were identified using RNA sequencing (RNA-Seq), verified with the real-time quantitative polymerase chain reaction (RT-qPCR). The functional-enrichment analysis including Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and Reactome enrichment analysis were performed to reveal the functional characteristics and related biological pathways of DEGs. Immunohistofluorescence (IHF) and flow cytometry (FCM) were used to detect immune cell subsets and proliferation markers. Results EPO treatment resulted in splenomegaly, spleen microstructure disorder, splenic corpuscular atrophy, indistinct germinal center, and unclear boundary between white and red pulp structures. RNA-Seq showed that EPO treatment suppressed gene expression associated with immune responses, while promoted cell cycle and DNA replication. IHF and FCM validated that, at the cellular level, T, B, M1 cells were significantly reduced, and M2 cells were significantly decreased after EPO treatment. The proliferation analysis showed that the portion of EDU+ or Ki-67+cells consisted of granulocytes and macrophages, and after EPO treatment, only macrophages showed a significant increase in their number and proportion, while granulocytes did not show a significant response to EPO stimulation. Conclusion Long term high-dose EPO treatment may lead to splenomegaly and immunosuppression of the local immune microenvironment in mice. The mechanism may be related to the increased anti-inflammatory and immunomodulatory functions caused by M2 cells. The study provides, for the first time, the transcriptomic characteristics and immunological of the spleens of EPO treated mice, providing a new perspective for the study of the effects of EPO on mice.
Collapse
Affiliation(s)
- Xinyi Lyu
- The Second Affiliated Hospital, Department of Vascular Surgery, Hengyang Medical School, University of South China, Hengyang, China
| | - Jiahao Shi
- The Second Affiliated Hospital, Department of Vascular Surgery, Hengyang Medical School, University of South China, Hengyang, China
| | - Qi Liu
- The Second Affiliated Hospital, Department of Vascular Surgery, Hengyang Medical School, University of South China, Hengyang, China
| | - Mingjun Jiang
- The Second Affiliated Hospital, Department of Vascular Surgery, Hengyang Medical School, University of South China, Hengyang, China
| | - Xilian Liu
- The Second Affiliated Hospital, Department of Endocrinology and Metabolism, Hengyang Medical School, University of South China, Hengyang, China
| | - Yulan Li
- The Second Affiliated Hospital, Department of Endocrinology and Metabolism, Hengyang Medical School, University of South China, Hengyang, China
| | - Shuqin Ding
- Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Xianpeng Dai
- The Second Affiliated Hospital, Department of Vascular Surgery, Hengyang Medical School, University of South China, Hengyang, China
| |
Collapse
|
8
|
Qiao T, Liu N, Cui Y, Gao Y, Lu J. A Review on the Potential Therapeutic Application of Macrophage Polarization in Recurrent Spontaneous Abortion; With an Emphasis on Natural Components. TOHOKU J EXP MED 2025; 265:47-58. [PMID: 39048350 DOI: 10.1620/tjem.2024.j069] [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: 07/27/2024]
Abstract
Three or more consecutive spontaneous pregnancy losses are the hallmark of recurrent spontaneous abortion (RSA) as a complex challenge in reproductive health, requiring a comprehensive understanding of contributing factors. Since balanced immune responses are essential for a successful pregnancy. Disruptions in immune responses may be the cause of unfavorable pregnancy outcomes like RSA. Of Note, Following RSA, immunopathological assessment of the placental implantation site markedly showed decidual inflammation, leading to hypothesize that RSA is a pregnancy disorder with an inflammatory etiology. Thus, an in-depth knowledge of how immune cells contribute to inflammation, may lead to the discovery of the novel therapeutic approaches for the prevention and/or treatment of RSA. Numerous studies have investigated the relationships between RSA and different immune cells, including B cells, T cells, decidual dendritic cells, and macrophages. Macrophages are present at the fetomaternal interface throughout pregnancy, and they are beneficial to the processes of embryonic development, placental formation, embryo implantation, and delivery. Macrophages classified as typically activated (M1, with the inflammatory role), or alternatively activated (M2, characterized by the anti-inflammatory role). Plants have a rich supply of strong bioactive components that can polarize macrophages toward an M1 pro-inflammatory state or an M2 anti-inflammatory phenotype. This review focuses on the potential role of derived plant-natural components in influencing macrophage polarization resulting in the management treatment of RSA.
Collapse
Affiliation(s)
- Tianqi Qiao
- Graduate School, Hebei North University
- Department of Reproductive Medicine, Handan Central Hospital
| | - Ningning Liu
- Department of gynecology, Handan Maternal and Child Health Hospital
| | - Yijian Cui
- Graduate School, Shenyang Medical College
- Department of Clinical Laboratory, Handan Central Hospital
| | - Yanxia Gao
- Department of Clinical Laboratory, Handan Maternal and Child Health Hospital
| | - Jing Lu
- Department of Reproductive Medicine, Handan Central Hospital
| |
Collapse
|
9
|
Astaneh ME, Fereydouni N. Nanocurcumin-enhanced zein nanofibers: Advancing macrophage polarization and accelerating wound healing. Regen Ther 2025; 28:51-62. [PMID: 39687330 PMCID: PMC11647652 DOI: 10.1016/j.reth.2024.11.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Revised: 11/10/2024] [Accepted: 11/20/2024] [Indexed: 12/18/2024] Open
Abstract
Introduction Chronic wounds continue to pose a significant global challenge, incurring substantial costs and necessitating extensive research in wound healing. Our previous work involved synthesizing zein nanofibers embedded with 5 %, 10 %, and 15 % nano-curcumin (Zein/nCUR 5, 10, and 15 % NFs), and examining their physicochemical and biological properties. This study aims to explore the potential of these nanofibers in macrophage (MØ) polarization and wound healing. Methods We assessed the survival of RAW264.7 cells cultured on Zein/nCUR 5, 10, and 15 % NFs using the MTT assay. To evaluate MØ polarization, we measured the expression of iNOS and Arg-1 genes in MØs cultured on Zein/nCUR 10 % NFs through real-time PCR. Furthermore, we examined the nanofibers' impact on pro-inflammatory cytokine expression (IL-1β, IL-6, TNF-α) in MØs via real-time PCR. The wound healing efficacy of Zein/nCUR 10 % NFs was tested on 54 male rats with full-thickness wounds, with assessments conducted on days 3, 7, and 14. Wound closure, re-epithelialization, and collagen secretion were evaluated through photographic analysis and tissue staining. Statistical analyses were performed using GraphPad Prism 6, with significance set at p < 0.05. Results Zein/nCUR 10 % NFs significantly enhanced the survival of RAW264.7 cells compared to other groups. They also markedly reduced iNOS expression and increased Arg-1 expression, indicating successful polarization of M1 to M2 MØs. Additionally, these nanofibers decreased the expression of IL-1β, IL-6, and TNF-α, and significantly improved wound closure, re-epithelialization, and collagen deposition compared to control and Zein groups. Conclusions This study demonstrates that Zein/nCUR 10 % NFs effectively polarize MØs from M1 to M2, significantly enhancing wound healing, thus offering a promising therapeutic approach for improved wound care.
Collapse
Affiliation(s)
- Mohammad Ebrahim Astaneh
- Department of Anatomical Sciences, School of Medicine, Fasa University of Medical Sciences, Fasa, Iran
- Department of Tissue Engineering, School of Medicine, Fasa University of Medical Sciences, Fasa, Iran
- Student Research Committee, Fasa University of Medical Sciences, Fasa, Iran
| | - Narges Fereydouni
- Department of Tissue Engineering, School of Medicine, Fasa University of Medical Sciences, Fasa, Iran
- Student Research Committee, Fasa University of Medical Sciences, Fasa, Iran
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| |
Collapse
|
10
|
Mohanty A, Vekariya V, Yadav S, Agrawal-Rajput R. Natural phytochemicals reverting M2 to M1 macrophages: A novel alternative leishmaniasis therapy. Microb Pathog 2025; 200:107311. [PMID: 39863089 DOI: 10.1016/j.micpath.2025.107311] [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: 05/14/2024] [Revised: 12/29/2024] [Accepted: 01/18/2025] [Indexed: 01/27/2025]
Abstract
INTRODUCTION Leishmaniasis is a tropical parasitic disease caused by the protozoan Leishmania which remains a significant global health concern with diverse clinical manifestations. Transmitted through the bite of an infected sandfly, its progression depends on the interplay between the host immune response and the parasite. The disease outcome is linked to macrophage polarisation into M1 and M2 phenotypes. M1 macrophages are pro-inflammatory and promote parasite clearance, while M2 macrophages support tissue repair and parasite survival by facilitating promastigote entry and intracellular amastigote proliferation. PURPOSE The review focuses on discovering novel phytochemicals that exploit the immunomodulatory properties of macrophages, which can serve as an alternative antileishmanial treatments due to their diverse chemical structures and ability to modulate immune responses. It examines the immunomodulatory effects of phytochemicals that directly or indirectly promote antileishmanial activity by influencing macrophage polarisation and cytokine secretion. They can induce M1 macrophage polarisation to directly combat leishmaniasis or suppress M2 macrophages, thereby exerting indirect antileishmanial activity by influencing the release of M1-and M2-related cytokines. RESULTS & DISCUSSION Phytochemicals demonstrate antileishmanial effects through ROS production, M1 activation, and cytokine modulation. They regulate M1/M2-related cytokines and macrophage activity, influencing immune responses. Although their effects may be non-specific, targeted delivery strategies could overcome current therapeutic limitations, positioning phytochemicals as promising candidates for leishmaniasis treatment to counter the limitations of current medications.
Collapse
Affiliation(s)
- Aditya Mohanty
- Immunology Lab, Biotechnology & Bioengineering, Indian Institute of Advanced Research, Gandhinagar, Gujarat, 382426, India
| | - Vasu Vekariya
- Immunology Lab, Biotechnology & Bioengineering, Indian Institute of Advanced Research, Gandhinagar, Gujarat, 382426, India
| | - Shivani Yadav
- Immunology Lab, Biotechnology & Bioengineering, Indian Institute of Advanced Research, Gandhinagar, Gujarat, 382426, India
| | - Reena Agrawal-Rajput
- Immunology Lab, Biotechnology & Bioengineering, Indian Institute of Advanced Research, Gandhinagar, Gujarat, 382426, India.
| |
Collapse
|
11
|
Yang L, Hou H, Lu L, Sun Y, Chen R, Deng Q, Chen H. Effects of natural source polysaccharides on neurological diseases: A review. Int J Biol Macromol 2025; 296:139697. [PMID: 39805435 DOI: 10.1016/j.ijbiomac.2025.139697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 01/05/2025] [Accepted: 01/07/2025] [Indexed: 01/16/2025]
Abstract
With the aging of society and changes in lifestyle, the incidence of neurological diseases (NDs) has been increasing year by year, bringing a heavy burden to patients and society. Although the efficacy of chemical drugs in the treatment of NDs is remarkable, there are problems such as high side effects and high costs. Therefore, finding mild and efficient drugs for NDs treatment has become an urgent clinical need. Natural source polysaccharides (NSPs) are macromolecules with unique bioactivity and low toxicity characteristics, which have great potential to become novel therapeutic agents for NDs. In the present study, the pharmacological activities and potential molecular mechanisms of NSPs to alleviate NDs are systematically reviewed from the perspectives of inflammation, oxidative stress, apoptosis, neuronal cell autophagy, neurotoxicity, and sedation-hypnosis. In addition, the limitations of the existing studies were analyzed and discussed, and the future research direction was suggested. This study may provide scientific basis for the research and development of therapeutic agents for NDs based on NSPs.
Collapse
Affiliation(s)
- Luyuan Yang
- Key Laboratory for Information System of Mountainous Areas and Protection of Ecological Environment, Guizhou Normal University, Guiyang 550001, China; Guizhou Engineering Laboratory for Quality Control & Evaluation Technology of Medicine, Guizhou Normal University, Guiyang 550001, China
| | - Hailu Hou
- Key Laboratory for Information System of Mountainous Areas and Protection of Ecological Environment, Guizhou Normal University, Guiyang 550001, China; Guizhou Engineering Laboratory for Quality Control & Evaluation Technology of Medicine, Guizhou Normal University, Guiyang 550001, China
| | - Liping Lu
- Guizhou Dalong Pharmaceutical Co., Ltd., Guiyang 550001, China
| | - Yu Sun
- Key Laboratory for Information System of Mountainous Areas and Protection of Ecological Environment, Guizhou Normal University, Guiyang 550001, China; Guizhou Engineering Laboratory for Quality Control & Evaluation Technology of Medicine, Guizhou Normal University, Guiyang 550001, China
| | - Ruhai Chen
- Key Laboratory for Information System of Mountainous Areas and Protection of Ecological Environment, Guizhou Normal University, Guiyang 550001, China; Guizhou Engineering Laboratory for Quality Control & Evaluation Technology of Medicine, Guizhou Normal University, Guiyang 550001, China
| | - Qingfang Deng
- Key Laboratory for Information System of Mountainous Areas and Protection of Ecological Environment, Guizhou Normal University, Guiyang 550001, China; Guizhou Engineering Laboratory for Quality Control & Evaluation Technology of Medicine, Guizhou Normal University, Guiyang 550001, China
| | - Huaguo Chen
- Key Laboratory for Information System of Mountainous Areas and Protection of Ecological Environment, Guizhou Normal University, Guiyang 550001, China; Guizhou Engineering Laboratory for Quality Control & Evaluation Technology of Medicine, Guizhou Normal University, Guiyang 550001, China.
| |
Collapse
|
12
|
Guy MM, Bian T, Sun L, Hao Y, Jiao X, Zhang W, Zhang T, Cui B. SLC7A11 is a potential therapeutic target and prognostic biomarker correlated with immune cell infiltration in cervical cancer. Discov Oncol 2025; 16:125. [PMID: 39915437 PMCID: PMC11802985 DOI: 10.1007/s12672-025-01888-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 02/03/2025] [Indexed: 02/09/2025] Open
Abstract
BACKGROUND SLC7A11 is importantly in both ferroptosis and disulfidptosis which participated in human development and homeostasis. By utilizing bioinformatics and in vitro validation, we explored SLC7A11's role in cervical cancer. METHODS From the TCGA database, we analyzed SLC7A11 expression profiles and validated its promoting role in cervical cancer by in vitro. Patients were divided into two subgroups according to SLC7A11 expression levels, and differentially expressed genes (DEGs) were identified between these groups. Then SLC7A11's mechanism was then clarified by function enrichment analysis. The association between SLC7A11 and the immune microenvironment was investigated. Finally, we explore potential drugs by oncoPredict. RESULTS Our findings suggest that SLC7A11 could serve as a valuable prognostic biomarker in cervical cancer. Besides, SLC7A11 was positively regulated cervical cancer cells' proliferation, migration and invasion. We identified 113 DEGs between two subgroups. Functional enrichment analysis revealed that these DEGs are linked to immune-related pathways. The SLC7A11 high expression group showed greater enrichment of resting NK cells, neutrophils, M0 macrophages, and activated mast cells, whereas the SLC7A11 low expression group had higher levels of resting dendritic cells, resting mast cells, and follicular helper T cells. Besides, there were higher TMB scores in patients with high SLC7A11 expression. Finally, we explore 37 kinds of drugs may improve prognosis. CONCLUSION SLC7A11, correlated with immune pathway, is a risk factor affecting the prognosis of cervical cancer. We also explore 37 kinds of drugs that may benefit cervical cancer patients.
Collapse
Affiliation(s)
- Mutangala Muloye Guy
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
- Department of Obstetrics and Gynecology, School of Medicine, University of Lubumbashi, Lubumbashi, Democratic Republic of the Congo
| | - Tingting Bian
- Department of Medical Imaging, Affiliated Hospital of Jining Medical University, Jining City, China
| | - Longyun Sun
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Yiping Hao
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Xinlin Jiao
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Wenjing Zhang
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Teng Zhang
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China.
| | - Baoxia Cui
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China.
| |
Collapse
|
13
|
Goleij P, Khandan M, Khazeei Tabari MA, Sanaye PM, Alijanzadeh D, Soltani A, Hosseini Z, Larsen DS, Khan H, Kumar AP, Daglia M. Unlocking the Potential: How Flavonoids Affect Angiogenesis, Oxidative Stress, Inflammation, Proliferation, Invasion, and Alter Receptor Interactions in Endometriosis. Food Sci Nutr 2025; 13:e4607. [PMID: 39803270 PMCID: PMC11716992 DOI: 10.1002/fsn3.4607] [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: 06/16/2024] [Revised: 10/19/2024] [Accepted: 10/27/2024] [Indexed: 01/16/2025] Open
Abstract
Endometriosis, though not classified as a carcinogenic condition, shares features such as oxidative stress, migration, invasion, angiogenesis, and inflammation with tumor cells. This study aims to review the effects of flavonoids on these processes and their molecular mechanisms in preventing and treating endometriosis. A comprehensive review was conducted, involving a literature search in online databases using keywords like "endometriosis," "endometrioma," and "flavonoid." Two authors screened the literature based on predefined criteria, and the selected studies were summarized in a structured data extraction table. Studies reviewed showed that various flavonoids impact key processes in endometriosis, including angiogenesis, inflammation, oxidative stress, and invasiveness. Flavonoids such as 2',7'-dichlorodihydrofluorescein diacetate (H2DCF-DA), naringenin, apigenin, myricetin, 5,7-dimethoxyflavone (DMF), chrysin, and 6,8-diprenylorobol were found to induce oxidative stress. Xanthohumol, isoliquiritigenin, and luteolin demonstrated effects on angiogenesis. Apigenin, isoliquiritigenin, and luteolin exhibited anti-inflammatory properties. Additionally, 3,6-dihydroxyflavone, isoliquiritigenin, and naringenin displayed anti-invasive activities. Flavonoid-receptor interactions further enhance their therapeutic potential in endometriosis management. Flavonoids such as nobiletin, chrysin, and daidzein modulate PPARγ and PPARα, reducing inflammation, promoting apoptosis, and improving lipid metabolism. These interactions regulate critical pathways in angiogenesis and immune responses. Additionally, flavonoids impact the aryl hydrocarbon receptor (AhR), with compounds like resveratrol inhibiting cell proliferation and cholesterol biosynthesis, further suppressing lesion growth. The ability of flavonoids like quercetin and kaempferol to antagonize NR4A1 leads to reduced cell proliferation and oxidative stress in endometriotic tissues. These findings offer insights into the mechanisms through which specific flavonoids modulate angiogenesis, inflammation, oxidative stress, and invasiveness in endometriosis. By targeting receptors such as PPARs, AhR, and NR4A1, flavonoids demonstrate the capacity to modulate both metabolic and inflammatory pathways, offering a multifaceted approach to managing endometriosis. Flavonoids can selectively target pathophysiologic molecules and pathways implicated in the condition. Consequently, leveraging the therapeutic attributes of flavonoids could lead to novel strategies for managing endometriosis.
Collapse
Affiliation(s)
- Pouya Goleij
- USERN OfficeKermanshah University of Medical SciencesKermanshahIran
- Department of Genetics, Faculty of BiologySana Institute of Higher EducationSariIran
- PhytoPharmacology Interest Group (PPIG)Universal Scientific Education and Research, Network (USERN)TehranIran
| | - Mohanna Khandan
- Student Research CommitteeMazandaran University of Medical SciencesSariIran
- USERN OfficeMazandaran University of Medical SciencesSariIran
| | - Mohammad Amin Khazeei Tabari
- Student Research CommitteeMazandaran University of Medical SciencesSariIran
- USERN OfficeMazandaran University of Medical SciencesSariIran
| | - Pantea Majma Sanaye
- PhytoPharmacology Interest Group (PPIG)Universal Scientific Education and Research, Network (USERN)TehranIran
- School of PharmacyZanjan University of Medical SciencesZanjanIran
| | - Dorsa Alijanzadeh
- Student Research CommitteeShahid Beheshti University of Medical SciencesTehranIran
- USERN OfficeShahid Beheshti University of Medical SciencesTehranIran
| | - Afsaneh Soltani
- Student Research CommitteeShahid Beheshti University of Medical SciencesTehranIran
- USERN OfficeShahid Beheshti University of Medical SciencesTehranIran
| | - Zahra Hosseini
- Student Research CommitteeMazandaran University of Medical SciencesSariIran
- USERN OfficeMazandaran University of Medical SciencesSariIran
| | - Danaé S. Larsen
- School of Chemical SciencesThe University of AucklandAucklandNew Zealand
| | - Haroon Khan
- Department of Pharmacy, Faculty of Chemical and Life SciencesAbdul Wali Khan University MardanMardanPakistan
- Department of PharmacyKorea UniversitySejongSouth Korea
| | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Maria Daglia
- Department of PharmacyUniversity of Naples “Federico II”NaplesItaly
- International Research Center for Food Nutrition and SafetyJiangsu UniversityZhenjiangChina
| |
Collapse
|
14
|
Du Q, Chen B, Yang X, Zhu H, Shams Ul Hassan S, Liu Q. Bioactive Macromolecule-mediated Biogenic FeONPs Attenuate Inflammation in Atherosclerotic Rat by Activating PI3K/Akt/eNOS Pathway. Curr Pharm Des 2025; 31:843-854. [PMID: 39317998 DOI: 10.2174/0113816128298009240828062231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 05/15/2024] [Accepted: 05/29/2024] [Indexed: 09/26/2024]
Abstract
INTRODUCTION Atherosclerosis refers to the thickening and hardening of artery walls. In our latest experiment, we utilized environmentally friendly techniques to produce multifunctional iron oxide nanoparticles (FeONPs) aimed at reducing inflammation in rats with atherosclerosis. METHODS The formulation was synthesized using curcumin (as the potent bioactive molecule) and was characterized. We assessed the in vitro antioxidant capability of the formulation against DPPH free radicals. Additionally, we quantified the mRNA levels of eNOS, PI3K, and Akt using Real Time-Polymerase Chain Reaction (RT-PCR). We tested the therapeutic impact of the bioactive formulation on a Triton X-100-induced atherosclerosis mouse model. RESULTS The crystallinity and magnetic behavior confirmed the magnetic properties of the FeONPs. The DPPH assay exhibited the dose-dependent radical scavenging characteristics of FeONPs. In the animal experiments, significant upregulation of the studied genes was noticed in treated groups 2 and 3 compared to treated group 1. Moreover, the expression of PI3K/eNOS/Akt was greater in treated group 3 than in treated group 2. These results indicate a dose-dependent elevation in target gene expression. CONCLUSION Nevertheless, the variation in gene expression between the negative control and the untreated control was not statistically significant (p > 0.05) across all genes.
Collapse
Affiliation(s)
- Qing Du
- Department of Emergency, The Fourth People's Hospital of Jinan, Shifan Road, No. 50, Jinan 250031, China
| | - Bo Chen
- Department of Cardiology, The Fourth People's Hospital of Jinan, Shifan Road, No. 50, Jinan 250031, China
| | - Xiaohan Yang
- Department of Cardiology, The Fourth People's Hospital of Jinan, Shifan Road, No. 50, Jinan 250031, China
| | - Hecheng Zhu
- Department of Cardiology, The Fourth People's Hospital of Jinan, Shifan Road, No. 50, Jinan 250031, China
| | | | - Qiang Liu
- Department of Cardiology, The Fourth People's Hospital of Jinan, Shifan Road, No. 50, Jinan 250031, China
| |
Collapse
|
15
|
Wang X, Zhang Y. Multi-omics joint screening of biomarkers related to M2 macrophages in gastric cancer. Discov Oncol 2024; 15:738. [PMID: 39623254 PMCID: PMC11612128 DOI: 10.1007/s12672-024-01623-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 11/25/2024] [Indexed: 12/06/2024] Open
Abstract
BACKGROUND Due to high mortality rate and limited treatments in gastric cancer (GC), call for deeper exploration of M2 macrophages as biomarkers is needed. METHODS The data for this study were obtained from the Gene Expression Omnibus (GEO) and Genomic Data Commons (GDC). The Seurat package was utilized for single-cell RNA sequencing (scRNA-seq) analysis. FindAllMarkers was used to identify genes highly expressed among different cell subsets. DESeq2 package was leveraged to screen differentially expressed genes (DEGs), while limma package was utilized for identifying differentially expressed proteins (DEPs). Enrichment analyses of the genes were conducted using KOBAS-i database. MultipleROC was applied to evaluate the diagnostic potential of biomarkers, and rms package was utilized to construct diagnostic models. hTFtarget database was utilized to predict potential transcription factors (TFs). Finally, cell-based assays were performed to validate the expression and potential biological functions of the screened key markers. RESULTS This study found that M2 macrophages were enriched in protein, endoplasmic reticulum, and virus-related pathways. A total of 4146 DEGs and 1946 DEPs were obtained through screening, with 254 common DEGs/DEPs. The results of gene function enrichment analysis suggested that it may affect the occurrence and development of GC through DNA replication and cell cycle. This study identified three biomarkers, HSPH1, HSPD1, and IFI30, and constructed a diagnostic model based on these three genes. The AUC value greater than 0.8 proved the reliability of the model. Through screening TFs, SPI1 and KLF5 were found to be the common TFs for the three biomarkers. The expression of the three genes IFI30, HSPD1 and HSPH1 was up-regulated in GC cells, and IFI30 may play a facilitating role in the migration and invasion of GC cells. CONCLUSION This study identified three biomarkers and constructed a diagnostic model, providing a new perspective for the research and treatment of GC.
Collapse
Affiliation(s)
- Xilong Wang
- Tumor Hematology Department, Liaoyang Central Hospital, Liaoyang, 111000, China
| | - Ying Zhang
- General Surgery Department, Liaoyang Central Hospital, Liaoyang, 111000, China.
| |
Collapse
|
16
|
Beigoli S, Boskabady MH. The molecular basis of the immunomodulatory effects of natural products: A comprehensive review. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156028. [PMID: 39276685 DOI: 10.1016/j.phymed.2024.156028] [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: 01/29/2024] [Revised: 07/21/2024] [Accepted: 09/02/2024] [Indexed: 09/17/2024]
Abstract
BACKGROUND Natural products (NPs) have long been recognized for their potential to modulate the immune system, offering a natural and holistic approach to enhancing immune function. In recent years, the immunomodulation effects of various natural products have attained significant attention. PURPOSE This article provides an overview of the role of natural products in immunomodulation, exploring their mechanisms of action, common types of NPs with immunomodulation properties, clinical applications, as well as considerations for their safety and efficacy. METHODS Extensive research has been conducted to compile important discoveries on the immunomodulatory properties of NPs through thorough searches of multiple databases such as PubMed, Science Direct, and Scopus up until January 2024. RESULTS By decreasing the levels of Th2 cytokines and pro-inflammatory cytokines, the results suggested that NPs have the ability to modulate the immune system. Therefore, NPs alleviate inflammation in various disorders such as asthma and cancer. Furthermore, the observed increase in CD4 cells and IFN-ɣ/IL4 levels, along with an increased IFN-c/IL4 ratio, indicates a stimulatory effect of NPs on Th1 activity in various inflammatory conditions. Therefore, NPs regulate the immune system by inhibiting T-cells and decreasing the growth of young B-cell lymphoma cells. CONCLUSION Reviewing studies indicated that NPs have the potential to serve as immunomodulatory candidates for treating disorders characterized by immune dysregulation. However, additional experimental and clinical studies are necessary before these agents can be implemented in clinical settings.
Collapse
Affiliation(s)
- Sima Beigoli
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Hossein Boskabady
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
17
|
Goleij P, Sanaye PM, Alam W, Zhang J, Tabari MAK, Filosa R, Jeandet P, Cheang WS, Efferth T, Khan H. Unlocking daidzein's healing power: Present applications and future possibilities in phytomedicine. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 134:155949. [PMID: 39217652 DOI: 10.1016/j.phymed.2024.155949] [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: 04/08/2023] [Revised: 07/29/2024] [Accepted: 08/08/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Cancer is one of the leading causes of death and a great threat to people around the world. Cancer treatment modalities include surgery, radiotherapy, chemotherapy, radiochemotherapy, hormone therapy, and immunotherapy. The best approach is to use a combination of several types. Among the treatment methods mentioned above, chemotherapy is frequently used, but its activity is hampered by the development of drug resistance and many side effects. In this regard, the use of medicinal plants has been discussed, and in recent decades, the use of isolated phytochemicals came into the focus of interest. By critically evaluating the available evidence and emphasizing the unique perspective offered by this review, we provide insights into the potential of daidzein as a promising therapeutic agent, as well as outline future research directions to optimize its efficacy in clinical settings. PURPOSE To summarized the therapeutic potential of daidzein, an isoflavone phytoestrogen in the management of several human diseases with the focuses on the current status and future prospects as a therapeutic agent. METHODS Several search engines, including PubMed, GoogleScholar, and ScienceDirect, were used, with the search terms "daidzein", "daidzein therapeutic potential", or individual effects. The study included all peer-reviewed articles. However, the most recent publications were given priority. RESULTS Daidzein showed protective effects against malignant diseases such as breast cancer, prostate cancer but also non-malignant diseases such as diabetes, osteoporosis, and cardiovascular diseases. Daidzein activates multiple signaling pathways leading to cell cycle arrest and apoptosis as well as antioxidant and anti-metastatic effects in malignant cells. Moreover, the anticancer effects against different cancer cells were more prominent and discussed in detail. CONCLUSIONS In short, daidzein represents a promising compound for drug development. The comprehensive potential anticancer activities of daidzein through various molecular mechanisms and its therapeutic/clinical status required further detail studies.
Collapse
Affiliation(s)
- Pouya Goleij
- USERN Office, Kermanshah University of Medical Sciences, Kermanshah, Iran; Department of Genetics, Faculty of Biology, Sana Institute of Higher Education, Sari, Iran; PhytoPharmacology Interest Group (PPIG), Universal Scientific Education and Research, Network (USERN), Tehran, Iran.
| | - Pantea Majma Sanaye
- PhytoPharmacology Interest Group (PPIG), Universal Scientific Education and Research, Network (USERN), Tehran, Iran; School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Waqas Alam
- Department of Pharmacy, Faculty of Chemical and Life Sciences, Abdul Wali Khan University Mardan, Mardan 23200, Pakistan
| | - Junmin Zhang
- School of Pharmacy, State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Mohammad Amin Khazeei Tabari
- PhytoPharmacology Interest Group (PPIG), Universal Scientific Education and Research, Network (USERN), Tehran, Iran; Student Research Committee, School of Medicine, Mazandaran University of Medical Sciences, Mazandaran, Iran
| | - Rosanna Filosa
- Department of Science and Technology, University of Sannio, Benevento 82100, Italy
| | - Philippe Jeandet
- Département de Biologie et Biochimie Faculté des Sciences Exactes et Naturelles Université de Reims BP 1039 51687, Reims CEDEX 02, France
| | - Wai San Cheang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, China
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Mainz 55128, Germany
| | - Haroon Khan
- Department of Pharmacy, Faculty of Chemical and Life Sciences, Abdul Wali Khan University Mardan, Mardan 23200, Pakistan.
| |
Collapse
|
18
|
Shen LS, Lin Z, Gong RH, Lin YS, Qiao XF, Hu QM, Qin WH, Chen S, Yang Y, Chen GQ. β,β-Dimethylacrylalkannin, a Natural Naphthoquinone, Inhibits the Growth of Hepatocellular Carcinoma Cells by Modulating Tumor-Associated Macrophages. Molecules 2024; 29:3919. [PMID: 39202998 PMCID: PMC11357245 DOI: 10.3390/molecules29163919] [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: 07/03/2024] [Revised: 07/19/2024] [Accepted: 07/25/2024] [Indexed: 09/03/2024] Open
Abstract
Tumor-associated macrophages (TAMs) are pivotal in the tumor microenvironment (TME) of hepatocellular carcinoma (HCC), influencing various stages from initiation to metastasis. Understanding the role of TAMs in HCC is crucial for developing novel therapeutic strategies. Macrophages exhibit plasticity, resulting in M1 and M2 phenotypes, with M1 macrophages displaying antitumor properties and M2 macrophages promoting tumor progression. Targeting TAMs to alter their polarization could offer new avenues for HCC treatment. β,β-dimethylacrylalkannin (DMAKN), a natural naphthoquinone, has gained attention for its antitumor properties. However, its impact on TAMs modulation remains unclear. This study investigates DMAKN's modulation of TAMs and its anti-HCC activity. Using an in vitro model with THP-1 cells, we induced M1 macrophages with LPS/IFN-γ and M2 macrophages with IL-4/IL-13, confirming polarization with specific markers. Co-culturing these macrophages with HCC cells showed that M1 cells inhibited HCC growth, while M2 cells promoted it. Screening for non-toxic DMAKN concentrations revealed its ability to induce M1 polarization and enhance LPS/IFN-γ-induced M1 macrophages, both showing anti-HCC effects. Conversely, DMAKN suppressed IL-4/IL-13-induced M2 polarization, inhibiting M2 macrophages' promotion of HCC cell viability. In summary, DMAKN induces and enhances M1 polarization while inhibiting M2 polarization of macrophages, thereby inhibiting HCC cell growth. These findings suggest that DMAKN has the potential to regulate TAMs in HCC, offering promise for future therapeutic development.
Collapse
Affiliation(s)
- Li-Sha Shen
- Chongqing Academy of Chinese Materia Medica, Chongqing 400065, China (Y.Y.)
- Sichuan–Chongqing Joint Key Laboratory of Innovation of New Drugs of Traditional Chinese Medicine, Chongqing 400065, China
| | - Zesi Lin
- Southern Medical University of Hospital of Integrated Traditional Chinese Medicine and Western Medicine, Southern Medical University, Guangzhou 510515, China
| | - Rui-Hong Gong
- Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hung Hom, Hong Kong 999077, China (S.C.)
| | - Yu-Shan Lin
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518057, China
| | - Xing-Fang Qiao
- Chongqing Academy of Chinese Materia Medica, Chongqing 400065, China (Y.Y.)
- Sichuan–Chongqing Joint Key Laboratory of Innovation of New Drugs of Traditional Chinese Medicine, Chongqing 400065, China
| | - Qian-Mei Hu
- Chongqing Academy of Chinese Materia Medica, Chongqing 400065, China (Y.Y.)
- Sichuan–Chongqing Joint Key Laboratory of Innovation of New Drugs of Traditional Chinese Medicine, Chongqing 400065, China
| | - Wei-Han Qin
- Chongqing Academy of Chinese Materia Medica, Chongqing 400065, China (Y.Y.)
- Sichuan–Chongqing Joint Key Laboratory of Innovation of New Drugs of Traditional Chinese Medicine, Chongqing 400065, China
| | - Sibao Chen
- Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hung Hom, Hong Kong 999077, China (S.C.)
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518057, China
- Research Centre for Chinese Medicine Innovation, The Hong Kong Polytechnic University, Hung Hom, Hong Kong 999077, China
| | - Yong Yang
- Chongqing Academy of Chinese Materia Medica, Chongqing 400065, China (Y.Y.)
- Sichuan–Chongqing Joint Key Laboratory of Innovation of New Drugs of Traditional Chinese Medicine, Chongqing 400065, China
| | - Guo-Qing Chen
- Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hung Hom, Hong Kong 999077, China (S.C.)
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518057, China
- Research Centre for Chinese Medicine Innovation, The Hong Kong Polytechnic University, Hung Hom, Hong Kong 999077, China
| |
Collapse
|
19
|
Deng T, Xu J, Wang Q, Wang X, Jiao Y, Cao X, Geng Q, Zhang M, Zhao L, Xiao C. Immunomodulatory effects of curcumin on macrophage polarization in rheumatoid arthritis. Front Pharmacol 2024; 15:1369337. [PMID: 38487171 PMCID: PMC10938599 DOI: 10.3389/fphar.2024.1369337] [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: 01/12/2024] [Accepted: 02/19/2024] [Indexed: 03/17/2024] Open
Abstract
Rheumatoid arthritis (RA) is a systemic autoimmune disease characterized by synovial inflammation, cartilage destruction, pannus formation and bone erosion. Various immune cells, including macrophages, are involved in RA pathogenesis. The heterogeneity and plasticity of macrophages render them pivotal regulators of both the induction and resolution of the inflammatory response. Predominantly, two different phenotypes of macrophages have been identified: classically activated M1 macrophages exacerbate inflammation via the production of cytokines, chemokines and other inflammatory mediators, while alternatively activated M2 macrophages inhibit inflammation and facilitate tissue repair. An imbalance in the M1/M2 macrophage ratio is critical during the initiation and progression of RA. Macrophage polarization is modulated by various transcription factors, epigenetic elements and metabolic reprogramming. Curcumin, an active component of turmeric, exhibits potent immunomodulatory effects and is administered in the treatment of multiple autoimmune diseases, including RA. The regulation of macrophage polarization and subsequent cytokine production as well as macrophage migration is involved in the mechanisms underlying the therapeutic effect of curcumin on RA. In this review, we summarize the underlying mechanisms by which curcumin modulates macrophage function and polarization in the context of RA to provide evidence for the clinical application of curcumin in RA treatment.
Collapse
Affiliation(s)
- Tingting Deng
- Institute of Clinical Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Jiahe Xu
- Institute of Clinical Medicine, China-Japan Friendship Hospital, Beijing, China
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, China
| | - Qiong Wang
- Institute of Clinical Medicine, China-Japan Friendship Hospital, Beijing, China
- China-Japan Friendship Hospital, Capital Medical University, Beijing, China
| | - Xing Wang
- Institute of Clinical Medicine, China-Japan Friendship Hospital, Beijing, China
- China-Japan Friendship Hospital, Capital Medical University, Beijing, China
| | - Yi Jiao
- Institute of Clinical Medicine, China-Japan Friendship Hospital, Beijing, China
- China-Japan Friendship Hospital, Capital Medical University, Beijing, China
| | - Xiaoxue Cao
- Institute of Clinical Medicine, China-Japan Friendship Hospital, Beijing, China
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, China
| | - Qishun Geng
- Institute of Clinical Medicine, China-Japan Friendship Hospital, Beijing, China
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, China
| | - Mengxiao Zhang
- Institute of Clinical Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Lu Zhao
- Institute of Clinical Medicine, China-Japan Friendship Hospital, Beijing, China
- China-Japan Friendship Hospital, Capital Medical University, Beijing, China
| | - Cheng Xiao
- Institute of Clinical Medicine, China-Japan Friendship Hospital, Beijing, China
- Department of Emergency, China-Japan Friendship Hospital, Beijing, China
| |
Collapse
|
20
|
Abd-Elhalem SS, Al-Doori MH, Hassen MT. Macrophage Polarization Towards M2 Phenotype by Curcuminoids Through NF-κB Pathway Inhibition in Adjuvant-Induced Arthritis. Int Immunopharmacol 2023; 119:110231. [PMID: 37130441 DOI: 10.1016/j.intimp.2023.110231] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 04/19/2023] [Accepted: 04/20/2023] [Indexed: 05/04/2023]
Abstract
Macrophage polarization is decisive for homeostasis maintenance and tissue repair. Anti-inflammatory properties of curcumin (CUR) have been demonstrated in several studies. It used in the treatment of bone disorders includingrheumatoid arthritis. The present study aims to explore the potential mechanisms of curcumin on macrophage polarization, expression, activation, and cytokine secretion in adjuvant-induced arthritis as well as its possible role in enhancing the therapeutic action of methotrexate (MTX) together with minimizing MTX initiated side-effects. Rats were divided into eight groups as follows; Control group, MTX group: was weekly injected with MTX, CUR group: was treated with a daily oral dose of curcumin, MTX + CUR group: was treated with both methotrexate and curcumin, Adjuvant arthritis group (AIA): was injected with complete Freund's adjuvant for arthritis induction, AIA/MTX group: arthritic rats treated with methotrexate, AIA/CUR group: arthritic rats treated with curcumin and AIA/MTX + CUR: arthritic rats treated with both methotrexate and curcumin. Paw swelling, haematological analysis, immunological studies, histological observations and quantitative immunohistochemical investigations were performed. The present results showed that treating arthritic rats with curcumin either alone or in combination with methotrexate resulted in amelioration in paws inflammation, growth rate, absolute and relative spleen weights, and haematological analyses. Antinuclear antibodies, IL-1β, IL-8, IL-10, NF-kB levels, and CD68 + joint expression were also ameliorated. The microscopic examination of joint and spleen showed more improvement as apparently normal tissues in treated groups. It can be concluded that curcumin seems to be most promising in regulating macrophage expression, activation, cytokine secretion, and polarization, thus providing a novel insight in the application of curcumin-based treatments.
Collapse
Affiliation(s)
- Sahar S Abd-Elhalem
- Zoology Department, Faculty of Women for Arts, Science and Education, Ain Shams University, 11757 Cairo, Egypt.
| | - Mohamed H Al-Doori
- Analysis Pathological Department, Faculty of Applied Sciences, Samarra University, Iraq
| | - Marwa T Hassen
- Zoology Department, Faculty of Women for Arts, Science and Education, Ain Shams University, 11757 Cairo, Egypt
| |
Collapse
|
21
|
Kassouri C, Rodriguez Torres S, Gonzalez Suarez N, Duhamel S, Annabi B. EGCG Prevents the Transcriptional Reprogramming of an Inflammatory and Immune-Suppressive Molecular Signature in Macrophage-like Differentiated Human HL60 Promyelocytic Leukemia Cells. Cancers (Basel) 2022; 14:5065. [PMID: 36291849 PMCID: PMC9599716 DOI: 10.3390/cancers14205065] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/07/2022] [Accepted: 10/13/2022] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND The promyelocytic leukemia cell differentiation process enables recapitulation of the polarized M1 or M2 macrophage-like phenotype with inflammatory and immune-suppressive properties. While evidence supports the anti-inflammatory effect of dietary-derived epigallocatechin-3-gallate (EGCG), its impact on the onset of immune phenotype molecular signature remains unclear. METHODS Human HL60 promyelocytic cells grown in suspension were differentiated into CD11bHigh/CD14Low adherent macrophages with phorbol 12-myristate 13-acetate (PMA). Gelatin zymography was used to assess the levels of matrix metalloproteinase (MMP)-9, and total RNA was isolated for RNAseq and RT-qPCR assessment of differentially expressed gene levels involved in inflammation and immunity. Protein lysates were used to assess the phosphorylation status of signaling intermediates involved in macrophage-like cell differentiation. RESULTS Cell adhesion and induction of MMP-9 were indicative of HL60 cell differentiation into a macrophage-like phenotype. The extracellular signal-regulated kinase (ERK), glycogen synthase kinase (GSK)-3, p90 ribosomal S6 kinases (RSK), and cAMP-response-element-binding protein (CREB) were all phosphorylated, and EGCG reduced such phosphorylation status. Increases in inflammation and immunity genes included, among others, CCL22, CSF1, CSF2, IL1B, and TNF, which inductions were prevented by EGCG. This was corroborated by unbiased transcriptomic analysis which further highlighted the capacity of EGCG to downregulate the hematopoietic stem cell regulator CBFA2T3. CONCLUSION EGCG inhibits inflammatory signaling crosstalk and prevents the onset of an immune phenotype in macrophage-like differentiated cells.
Collapse
Affiliation(s)
- Celia Kassouri
- Laboratoire d’Oncologie Moléculaire, Département de Chimie, and CERMO-FC, Université du Québec à Montréal, Montreal, QC H2X 2J6, Canada
| | - Sahily Rodriguez Torres
- Laboratoire d’Oncologie Moléculaire, Département de Chimie, and CERMO-FC, Université du Québec à Montréal, Montreal, QC H2X 2J6, Canada
| | - Narjara Gonzalez Suarez
- Laboratoire d’Oncologie Moléculaire, Département de Chimie, and CERMO-FC, Université du Québec à Montréal, Montreal, QC H2X 2J6, Canada
| | - Stéphanie Duhamel
- Goodman Cancer Institute, McGill University, Montreal, QC H3A 1A3, Canada
| | - Borhane Annabi
- Laboratoire d’Oncologie Moléculaire, Département de Chimie, and CERMO-FC, Université du Québec à Montréal, Montreal, QC H2X 2J6, Canada
| |
Collapse
|