1
|
Liu ZC, Fu HJ, Li NC, Deng FJ, Gan YK, Ye YJ, Huang BH, Liu C, Chen JH, Li XF. Systematic pharmacology and experimental validation to elucidate the inflammation-associated mechanism of Huanglian Wendan (HLWD) decoction in the treatment of MAFLD associated with atherosclerosis. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118841. [PMID: 39299361 DOI: 10.1016/j.jep.2024.118841] [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: 06/09/2024] [Revised: 09/14/2024] [Accepted: 09/16/2024] [Indexed: 09/22/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Metabolic-associated fatty liver disease (MAFLD) and atherosclerosis are very common disorders that frequently coexist. The therapeutic efficacy of Huanglian Wendan (HLWD) decoction, a traditional Chinese medicine (TCM) prescription, is satisfactory in treating MAFLD associated with atherosclerosis. However, the underlying mechanisms through which HLWD exerts its effects need to be elucidated. Given the complex composition of HLWD and its multiple therapeutic targets, pharmacological investigation is challenging. AIM OF THIS STUDY This study aimed to identify the effective compounds in HLWD and elucidate the mechanisms involved in its therapeutic effect on MAFLD associated with atherosclerosis. MATERIALS AND METHODS We used a systematic pharmacology method to identify effective compounds present in HLWD and determine the mechanism by which it affects MAFLD associated with atherosclerosis. The effective components of HLWD were identified through ultrahigh-performance liquid chromatography-q exactive-orbitrap high resolution mass spectrometry (UHPLC-Q-Orbitrap HRMS). Next, a comprehensive in silico method was used to predict potential related targets and disease targets for these compounds to establish corresponding pathways. The accuracy of our assumed systemic pharmacology results was determined by conducting follow-up experiments. RESULTS By conducting UHPLC-Q-Orbitrap HRMS combined with network analysis, we identified 18 potentially active components of HLWD and assessed the inflammatory regulatory mechanism by which it affects MAFLD associated with atherosclerosis on the basis of 52 key targets. We used a high-fat, high-cholesterol (HFHC)-induced mice model of MAFLD associated with atherosclerosis to confirm our results. We found that administering HLWD significantly improved the appearance of their liver and reduced their body weight, liver weight, blood lipids, hepatic damage, and hepatic pathology. HLWD also decreased atherosclerotic lesion areas, foam cells, and inflammatory cells in the aorta. HLWD showed anti-inflammatory effects, suppressed M1 polarization, and promoted M2 polarization in the liver and aorta. HLWD might also regulate peroxisome proliferator-activated receptor-γ (PPARγ)/nuclear factor kappa-B (NF-κB) signaling to influence macrophage polarization and inflammation. CONCLUSIONS Our results showed that HLWD protected against HFHC diet-induced MAFLD associated with atherosclerosis by regulating PPARγ/NF-κB signaling, thus adjusting macrophage polarization and inflammation. Additionally, pharmacochemistry research, network pharmacology analysis, and experimental verification can be combined to form a comprehensive model used in studies on TCM.
Collapse
Affiliation(s)
- Zhi-Chao Liu
- School of Rehabilitation Medicine, Shandong Second Medical University, Weifang, Shandong Province, 261053, PR China.
| | - Huan-Jie Fu
- Department of Cardiovascular, Second Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300150, PR China.
| | - Ning-Cen Li
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, PR China.
| | - Fang-Jun Deng
- Department of Cardiovascular, Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin, 300150, PR China.
| | - Yong-Kang Gan
- Department of Vascular Surgery, Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin, 300150, PR China.
| | - Yu-Jia Ye
- School of Rehabilitation Medicine, Shandong Second Medical University, Weifang, Shandong Province, 261053, PR China.
| | - Bing-Hui Huang
- School of Rehabilitation Medicine, Shandong Second Medical University, Weifang, Shandong Province, 261053, PR China.
| | - Chang Liu
- School of Rehabilitation Medicine, Shandong Second Medical University, Weifang, Shandong Province, 261053, PR China.
| | - Jin-Hong Chen
- School of Rehabilitation Medicine, Shandong Second Medical University, Weifang, Shandong Province, 261053, PR China.
| | - Xiao-Feng Li
- Department of Cardiovascular, Second Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300150, PR China.
| |
Collapse
|
2
|
Li J, Ma W, Tang Z, Li Y, Zheng R, Xie Y, Li G. Macrophage‑driven pathogenesis in acute lung injury/acute respiratory disease syndrome: Harnessing natural products for therapeutic interventions (Review). Mol Med Rep 2025; 31:16. [PMID: 39513609 PMCID: PMC11551695 DOI: 10.3892/mmr.2024.13381] [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: 05/15/2024] [Accepted: 09/27/2024] [Indexed: 11/15/2024] Open
Abstract
Acute lung injury (ALI) or acute respiratory distress syndrome (ARDS) is a common respiratory disease characterized by hypoxemia and respiratory distress. It is associated with high morbidity and mortality. Due to the complex pathogenesis of ALI, the clinical management of patients with ALI/ARDS is challenging, resulting in numerous post‑treatment sequelae and compromising the quality of life of patients. Macrophages, as a class of innate immune cells, play an important role in ALI/ARDS. In recent years, the functions and phenotypes of macrophages have been better understood due to the development of flow cytometry, immunofluorescence, single‑cell sequencing and spatial genomics. However, no macrophage‑targeted drugs for the treatment of ALI/ARDS currently exist in clinical practice. Natural products are important for drug development, and it has been shown that numerous natural compounds from herbal medicine can alleviate ALI/ARDS caused by various factors by modulating macrophage abnormalities. In the present review, the natural products from herbal medicine that can modulate macrophage abnormalities in ALI/ARDS to treat ALI/ARDS are introduced, and their mechanisms of action, discovered in the previous five years (2019‑2024), are presented. This will provide novel ideas and directions for further research, to develop new drugs for the treatment of ALI/ARDS.
Collapse
Affiliation(s)
- Jincun Li
- College of Traditional Chinese Medicine, Yunnan University of Chinese Medicine, Kunming, Yunnan 650500, P.R. China
| | - Wenyu Ma
- College of Traditional Chinese Medicine, Yunnan University of Chinese Medicine, Kunming, Yunnan 650500, P.R. China
| | - Zilei Tang
- College of Traditional Chinese Medicine, Yunnan University of Chinese Medicine, Kunming, Yunnan 650500, P.R. China
| | - Yingming Li
- College of Traditional Chinese Medicine, Yunnan University of Chinese Medicine, Kunming, Yunnan 650500, P.R. China
| | - Ruiyu Zheng
- College of Traditional Chinese Medicine, Yunnan University of Chinese Medicine, Kunming, Yunnan 650500, P.R. China
| | - Yuhuan Xie
- Yunnan Innovation Team of Application Research on Traditional Chinese Medicine Theory of Disease Prevention, Yunnan University of Chinese Medicine, Kunming, Yunnan 650500, P.R. China
- Yunnan Provincial University Key Laboratory of Aromatic Chinese Herb Research, Basic Medical School, Yunnan University of Chinese Medicine, Kunming, Yunnan 650500, P.R. China
| | - Gang Li
- Yunnan Provincial University Key Laboratory of Aromatic Chinese Herb Research, Basic Medical School, Yunnan University of Chinese Medicine, Kunming, Yunnan 650500, P.R. China
- Basic Medical School, Yunnan University of Chinese Medicine, Kunming, Yunnan 650500, P.R. China
| |
Collapse
|
3
|
Gudgeon J, Dannoura A, Chatterjee R, Sidgwick F, Raymond BB, Frey AM, Marin-Rubio JL, Trost M. Mass spectrometry-based proteomic exploration of diverse murine macrophage cellular models. Life Sci Alliance 2025; 8:e202402760. [PMID: 39510801 PMCID: PMC11544424 DOI: 10.26508/lsa.202402760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 10/18/2024] [Accepted: 10/18/2024] [Indexed: 11/15/2024] Open
Abstract
Immortalised cell lines that mimic their primary cell counterparts are fundamental to research, particularly when large cell numbers are required. Here, we report that immortalisation of bone marrow-derived macrophages (iBMDMs) using the J2 virus resulted in the loss of a protein of interest, MSR1, in WT cells by an unknown mechanism. This led us to perform an in-depth mass spectrometry-based proteomic characterisation of common murine macrophage cell lines (J774A.1, RAW264.7, and BMA3.1A7), in comparison with the iBMDMs, as well as primary BMDMs from both C57BL/6 and BALB/c mice. This analysis revealed striking differences in protein profiles associated with macrophage polarisation, phagocytosis, pathogen recognition, and interferon signalling. Among the cell lines, J774A.1 cells were the most similar to the gold standard primary BMDM model, whereas BMA3.1A7 cells were the least similar because of the reduction in abundance of several key proteins related closely to macrophage function. This comprehensive proteomic dataset offers valuable insights into the use and suitability of macrophage cell lines for cell signalling and inflammation research.
Collapse
Affiliation(s)
- Jack Gudgeon
- https://ror.org/01kj2bm70 Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Abeer Dannoura
- https://ror.org/01kj2bm70 Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Ritika Chatterjee
- https://ror.org/01kj2bm70 Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Frances Sidgwick
- https://ror.org/01kj2bm70 Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Benjamin Ba Raymond
- https://ror.org/01kj2bm70 Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Andrew M Frey
- https://ror.org/01kj2bm70 Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - José Luis Marin-Rubio
- https://ror.org/01kj2bm70 Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Matthias Trost
- https://ror.org/01kj2bm70 Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
4
|
Rughetti A, Bharti S, Savai R, Barmpoutsi S, Weigert A, Atre R, Siddiqi F, Sharma R, Khabiya R, Hirani N, Baig MS. Imperative role of adaptor proteins in macrophage toll-like receptor signaling pathways. Future Sci OA 2024; 10:2387961. [PMID: 39248050 PMCID: PMC11385170 DOI: 10.1080/20565623.2024.2387961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 07/30/2024] [Indexed: 09/10/2024] Open
Abstract
Macrophages are integral part of the body's defense against pathogens and serve as vital regulators of inflammation. Adaptor molecules, featuring diverse domains, intricately orchestrate the recruitment and transmission of inflammatory responses through signaling cascades. Key domains involved in macrophage polarization include Toll-like receptors (TLRs), Src Homology2 (SH2) and other small domains, alongside receptor tyrosine kinases, crucial for pathway activation. This review aims to elucidate the enigmatic role of macrophage adaptor molecules in modulating macrophage activation, emphasizing their diverse roles and potential therapeutic and investigative avenues for further exploration.
Collapse
Affiliation(s)
- Aurelia Rughetti
- Laboratory of Tumor Immunology & Cell Therapy, Department of Experimental Medicine, Policlinico Umberto I, University of Rome "Sapienza", Rome, Italy
| | - Shreya Bharti
- Department of Biosciences & Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Indore, India
| | - Rajkumar Savai
- Lung Microenvironmental Niche in Cancerogenesis, Institute for Lung Health (ILH), Justus Liebig University, Giessen, D-35390, Germany
- Max Planck Institute for Heart & Lung Research, Member of the German Center for Lung Research (DZL), Member of the Cardio-Pulmonary Institute (CPI), Bad Nauheim, D-61231, Germany
- Institute of Biochemistry, Faculty of Medicine, Goethe University Frankfurt, Frankfurt, D-60590, Germany
| | - Spyridoula Barmpoutsi
- Lung Microenvironmental Niche in Cancerogenesis, Institute for Lung Health (ILH), Justus Liebig University, Giessen, D-35390, Germany
- Max Planck Institute for Heart & Lung Research, Member of the German Center for Lung Research (DZL), Member of the Cardio-Pulmonary Institute (CPI), Bad Nauheim, D-61231, Germany
| | - Andreas Weigert
- Institute of Biochemistry, Faculty of Medicine, Goethe University Frankfurt, Frankfurt, D-60590, Germany
- Frankfurt Cancer Institute (FCI), Goethe University Frankfurt, Frankfurt, D-60323, Germany
| | - Rajat Atre
- Department of Biosciences & Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Indore, India
| | - Faaiza Siddiqi
- Department of Biosciences & Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Indore, India
| | - Rahul Sharma
- Department of Biosciences & Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Indore, India
| | - Rakhi Khabiya
- Department of Biosciences & Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Indore, India
| | - Nik Hirani
- MRC Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, EH164TJ, UK
| | - Mirza S Baig
- Department of Biosciences & Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Indore, India
| |
Collapse
|
5
|
Wan J, Yang F, Tong S, Zhou T, Wang S. Triggering receptor expressed on myeloid cells-1 aggravates obliterative bronchiolitis via enhancing the proinflammatory phenotype of macrophages. Int Immunopharmacol 2024; 143:113274. [PMID: 39353383 DOI: 10.1016/j.intimp.2024.113274] [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: 09/16/2024] [Accepted: 09/25/2024] [Indexed: 10/04/2024]
Abstract
BACKGROUND Triggering Receptor Expressed on Myeloid cells-1 (TREM-1) plays an important role in innate immune system. However, whether and how TREM-1 contributes to obliterative bronchiolitis (OB) progression remains unclear. METHODS A murine orthotopic tracheal transplantation model was constructed to mimic the pathogenesis of OB. qPCR and immunoblotting were used to measure TREM-1 expression. RNA sequencing was used to investigate the impact of TREM-1 on proinflammatory phenotype of macrophages. Trem-1 knockout mice and Nlrp3 knockout mice were generated to investigate the role of the TREM-1/NLRP3 pathway in the proinflammatory phenotype of macrophages. The infiltration of immune cells within the grafts was quantified using immunofluorescence staining. Flow cytometry was used to detect the proportion of different immune cells in mice spleen and the expression levels of iNOS and co-stimulatory molecules in macrophages. RESULTS The expression of TREM-1 was upregulated in the mouse OB model. Genetic ablation or pharmacological inhibition of TREM-1 ameliorated OB, whereas the stimulation of TREM-1 using anti-TREM-1 agonistic antibody exacerbated OB. Moreover, Trem-1 ablation reduced the infiltration of iNOS+ macrophages and limited the T cell responses. In vitro studies revealed that Trem-1 deletion impaired the proinflammatory function and antigen presentation ability of macrophages. Additionally, Trem-1 knockout inhibited the activation of NLRP3 signaling pathway. NLRP3 overexpression restored the proinflammatory phenotype of Trem-1 knockout macrophages. CONCLUSIONS These findings indicated that TREM-1 could promote the proinflammatory phenotype of macrophages through NLRP3 inflammasome activation, thereby exacerbating OB progression. These findings indicated that TREM-1 may serve as a therapeutic target for OB treatment.
Collapse
Affiliation(s)
- Junhao Wan
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Fengjing Yang
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Song Tong
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Ting Zhou
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Sihua Wang
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
6
|
Luo WJ, Dong XW, Ye H, Zhao QS, Zhang QB, Guo WY, Liu HW, Xu F. Vitamin D 1,25-Dihydroxyvitamin D 3 reduces lipid accumulation in hepatocytes by inhibiting M1 macrophage polarization. World J Gastrointest Oncol 2024; 16:4685-4699. [DOI: 10.4251/wjgo.v16.i12.4685] [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: 04/16/2024] [Revised: 09/09/2024] [Accepted: 10/08/2024] [Indexed: 11/12/2024] Open
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD), which is a significant liver condition associated with metabolic syndrome, is the leading cause of liver diseases globally and its prevalence is on the rise in most nations. The protective impact of vitamin D on NAFLD and its specific mechanism remains unclear.
AIM To examine the role of vitamin D in NAFLD and how vitamin D affects the polarization of hepatic macrophages in NAFLD through the vitamin D receptor (VDR)-peroxisome proliferator activated receptor (PPAR)γ pathway.
METHODS Wild-type C57BL/6 mice were provided with a high-fat diet to trigger NAFLD model and administered 1,25-dihydroxy-vitamin D [1,25(OH)2D3] supplementation. 1,25(OH)2D3 was given to RAW264.7 macrophages that had been treated with lipid, and a co-culture with AML12 hepatocytes was set up. Lipid accumulation, lipid metabolism enzymes, M1/M2 phenotype markers, proinflammatory cytokines and VDR-PPARγ pathway were determined.
RESULTS Supplementation with 1,25(OH)2D3 relieved hepatic steatosis and decreased the proinflammatory M1 polarization of hepatic macrophages in NAFLD. Administration of 1,25(OH)2D3 suppressed the proinflammatory M1 polarization of macrophages induced by fatty acids, thereby directly relieving lipid accumulation and metabolism in hepatocytes. The VDR-PPARγ pathway had a notable impact on reversing lipid-induced proinflammatory M1 polarization of macrophages regulated by the administration of 1,25(OH)2D3.
CONCLUSION Supplementation with 1,25(OH)2D3 improved hepatic steatosis and lipid metabolism in NAFLD, linked to its capacity to reverse the proinflammatory M1 polarization of hepatic macrophages, partially by regulating the VDR-PPARγ pathway. The involvement of 1,25(OH)2D3 in inhibiting fatty-acid-induced proinflammatory M1 polarization of macrophages played a direct role in relieving lipid accumulation and metabolism in hepatocytes.
Collapse
Affiliation(s)
- Wen-Jing Luo
- Department of Gastroenterology, Ningbo Medical Center Lihuili Hospital, Ningbo 315000, Zhejiang Province, China
| | - Xian-Wen Dong
- Department of Gastroenterology, Ningbo Medical Center Lihuili Hospital, Ningbo 315000, Zhejiang Province, China
| | - Hua Ye
- Department of Gastroenterology, Ningbo Medical Center Lihuili Hospital, Ningbo 315000, Zhejiang Province, China
| | - Qiao-Su Zhao
- Department of Gastroenterology, Ningbo Medical Center Lihuili Hospital, Ningbo 315000, Zhejiang Province, China
| | - Qiu-Bo Zhang
- Department of Gastroenterology, Ningbo Medical Center Lihuili Hospital, Ningbo 315000, Zhejiang Province, China
| | - Wen-Ying Guo
- Department of Gastroenterology, Ningbo Medical Center Lihuili Hospital, Ningbo 315000, Zhejiang Province, China
| | - Hui-Wei Liu
- Department of Gastroenterology, Ningbo Medical Center Lihuili Hospital, Ningbo 315000, Zhejiang Province, China
| | - Feng Xu
- Department of Gastroenterology, Ningbo Medical Center Lihuili Hospital, Ningbo 315000, Zhejiang Province, China
| |
Collapse
|
7
|
Huart C, Gupta MS, Van Ginderachter JA. The role of RNA modifications in disease-associated macrophages. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102315. [PMID: 39296330 PMCID: PMC11408368 DOI: 10.1016/j.omtn.2024.102315] [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] [Indexed: 09/21/2024]
Abstract
In recent years, the field of epitranscriptomics has witnessed significant breakthroughs with the identification of more than 150 different chemical modifications in different RNA species. It has become increasingly clear that these chemical modifications play an important role in the regulation of fundamental processes linked to cell fate and development. Further interest was sparked by the ability of the epitranscriptome to regulate pathogenesis. However, despite the involvement of macrophages in a multitude of diseases, a clear knowledge gap exists in the understanding of how RNA modifications regulate the phenotype of these cells. Here, we provide a comprehensive overview of the known roles of macrophage RNA modifications in the context of different diseases.
Collapse
Affiliation(s)
- Camille Huart
- Lab of Cellular and Molecular Immunology, Brussels Center for Immunology (BCIM), Vrije Universiteit Brussel, Brussels, Belgium
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium
| | - Mayuk Saibal Gupta
- Lab of Cellular and Molecular Immunology, Brussels Center for Immunology (BCIM), Vrije Universiteit Brussel, Brussels, Belgium
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium
| | - Jo A Van Ginderachter
- Lab of Cellular and Molecular Immunology, Brussels Center for Immunology (BCIM), Vrije Universiteit Brussel, Brussels, Belgium
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium
| |
Collapse
|
8
|
Wang X, Hu J, Xie S, Li W, Zhang H, Huang L, Qian Z, Zhao C, Zhang L. Hidden role of microglia during neurodegenerative disorders and neurocritical care: A mitochondrial perspective. Int Immunopharmacol 2024; 142:113024. [PMID: 39217875 DOI: 10.1016/j.intimp.2024.113024] [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: 05/07/2024] [Revised: 08/04/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024]
Abstract
The incidence of aging-related neurodegenerative disorders and neurocritical care diseases is increasing worldwide. Microglia, the main inflammatory cells in the brain, could be potential viable therapeutic targets for treating neurological diseases. Interestingly, mitochondrial functions, including energy metabolism, mitophagy and transfer, fission and fusion, and mitochondrial DNA expression, also change in activated microglia. Notably, mitochondria play an active and important role in the pathophysiology of neurodegenerative disorders and neurocritical care diseases. This review briefly summarizes the current knowledge on mitochondrial dysfunction in microglia in neurodegenerative disorders and neurocritical care diseases and comprehensively discusses the prospects of the application of neurological injury prevention and treatment targets by mitochondria.
Collapse
Affiliation(s)
- Xinrun Wang
- Department of Critical Care Medicine, Hunan Provincial Clinical Research Center for Critical Care Medicine, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China
| | - Jiyun Hu
- Department of Critical Care Medicine, Hunan Provincial Clinical Research Center for Critical Care Medicine, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China
| | - Shucai Xie
- Department of Critical Care Medicine, Hunan Provincial Clinical Research Center for Critical Care Medicine, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China
| | - Wenchao Li
- Department of Critical Care Medicine, Hunan Provincial Clinical Research Center for Critical Care Medicine, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China
| | - Haisong Zhang
- Department of Critical Care Medicine, Hunan Provincial Clinical Research Center for Critical Care Medicine, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China
| | - Li Huang
- Department of Critical Care Medicine, Hunan Provincial Clinical Research Center for Critical Care Medicine, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China
| | - Zhaoxin Qian
- Department of Critical Care Medicine, Hunan Provincial Clinical Research Center for Critical Care Medicine, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China
| | - Chunguang Zhao
- Department of Critical Care Medicine, Hunan Provincial Clinical Research Center for Critical Care Medicine, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China.
| | - Lina Zhang
- Department of Critical Care Medicine, Hunan Provincial Clinical Research Center for Critical Care Medicine, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China.
| |
Collapse
|
9
|
Chen H, Han Z, Ma Y, Meng Q. Advances in macrophage-derived exosomes as immunomodulators in disease progression and therapy. Int Immunopharmacol 2024; 142:113248. [PMID: 39321698 DOI: 10.1016/j.intimp.2024.113248] [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: 06/18/2024] [Revised: 09/20/2024] [Accepted: 09/20/2024] [Indexed: 09/27/2024]
Abstract
Most somatic cells secrete vesicles called exosomes, which contain a variety of biomolecules. Recent research indicates that macrophage-derived exosomes are strongly correlated with tumors, infectious diseases, chronic inflammation, and tissue fibrosis. Therefore, the purpose of this review is to delve into the mechanisms of pathological states and how macrophage-derived exosomes react to them. We also discuss the biological effects of exosomes and how they affect disease. In addition, we have examined the possible uses of exosomes in illness treatment, highlighting both the benefits and drawbacks of these applications.
Collapse
Affiliation(s)
- Huizhu Chen
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing Key Laboratory of Sports Injuries, Beijing 100191, China; Peking University First Hospital, Peking University Health Science Center, Beijing 100034, China; Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing 100191, China.
| | - Ziping Han
- Institute of Cerebrovascular Diseases Research and Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China.
| | - Yong Ma
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing Key Laboratory of Sports Injuries, Beijing 100191, China; Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing 100191, China.
| | - Qingyang Meng
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing Key Laboratory of Sports Injuries, Beijing 100191, China; Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing 100191, China.
| |
Collapse
|
10
|
Günther K, Nischang V, Cseresnyés Z, Krüger T, Sheta D, Abboud Z, Heinekamp T, Werner M, Kniemeyer O, Beilhack A, Figge MT, Brakhage AA, Werz O, Jordan PM. Aspergillus fumigatus-derived gliotoxin impacts innate immune cell activation through modulating lipid mediator production in macrophages. Immunology 2024; 173:748-767. [PMID: 39268960 DOI: 10.1111/imm.13857] [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: 03/15/2024] [Accepted: 08/20/2024] [Indexed: 09/15/2024] Open
Abstract
Gliotoxin (GT), a secondary metabolite and virulence factor of the fungal pathogen Aspergillus fumigatus, suppresses innate immunity and supports the suppression of host immune responses. Recently, we revealed that GT blocks the formation of the chemotactic lipid mediator leukotriene (LT)B4 in activated human neutrophils and monocytes, and in rodents in vivo, by directly inhibiting LTA4 hydrolase. Here, we elucidated the impact of GT on LTB4 biosynthesis and the entire lipid mediator networks in human M1- and M2-like monocyte-derived macrophages (MDMs) and in human tissue-resident alveolar macrophages. In activated M1-MDMs with high capacities to generate LTs, the formation of LTB4 was effectively suppressed by GT, connected to attenuated macrophage phagocytic activity as well as human neutrophil movement and migration. In resting macrophages, especially in M1-MDMs, GT elicited strong formation of prostaglandins, while bacterial exotoxins from Staphylococcus aureus evoked a broad spectrum of lipid mediator biosynthesis in both MDM phenotypes. We conclude that GT impairs functions of activated innate immune cells through selective suppression of LTB4 biosynthesis, while GT may also prime the immune system by provoking prostaglandin formation in macrophages.
Collapse
Affiliation(s)
- Kerstin Günther
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Jena, Germany
| | - Vivien Nischang
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Jena, Germany
| | - Zoltan Cseresnyés
- Applied Systems Biology, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll Institute (Leibniz-HKI), Jena, Germany
| | - Thomas Krüger
- Department of Molecular and Applied Microbiology, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knöll Institute (Leibniz-HKI), Jena, Germany
| | - Dalia Sheta
- Department of Internal Medicine II, University Hospital Würzburg, Center of Experimental Molecular Medicine, Würzburg, Germany
| | - Zahraa Abboud
- Department of Internal Medicine II, University Hospital Würzburg, Center of Experimental Molecular Medicine, Würzburg, Germany
| | - Thorsten Heinekamp
- Department of Molecular and Applied Microbiology, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knöll Institute (Leibniz-HKI), Jena, Germany
| | - Markus Werner
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Jena, Germany
| | - Olaf Kniemeyer
- Department of Molecular and Applied Microbiology, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knöll Institute (Leibniz-HKI), Jena, Germany
| | - Andreas Beilhack
- Department of Internal Medicine II, University Hospital Würzburg, Center of Experimental Molecular Medicine, Würzburg, Germany
| | - Marc Thilo Figge
- Applied Systems Biology, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll Institute (Leibniz-HKI), Jena, Germany
- Institute of Microbiology, Friedrich Schiller University Jena, Jena, Germany
| | - Axel A Brakhage
- Department of Molecular and Applied Microbiology, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knöll Institute (Leibniz-HKI), Jena, Germany
- Institute of Microbiology, Friedrich Schiller University Jena, Jena, Germany
| | - Oliver Werz
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Jena, Germany
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Jena, Germany
| | - Paul M Jordan
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Jena, Germany
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Jena, Germany
| |
Collapse
|
11
|
Gu Y, Yu S, Gu W, Li B, Xue J, Liu J, Zhang Q, Yin Y, Zhang H, Guo Q, Yuan M, Lyu Z, Mu Y, Cheng Y. M2 macrophage infusion ameliorates diabetic glomerulopathy via the JAK2/STAT3 pathway in db/db mice. Ren Fail 2024; 46:2378210. [PMID: 39090966 PMCID: PMC11299449 DOI: 10.1080/0886022x.2024.2378210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 07/02/2024] [Accepted: 07/04/2024] [Indexed: 08/04/2024] Open
Abstract
Objectives: To explore the therapeutic effects of M2 macrophages in diabetic nephropathy (DN) and their mechanism.Methods: We infused M2 macrophages stimulated with IL-4 into 10-week-old db/db mice once a week for 4 weeks through the tail vein as M2 group. Then we investigated the role of M2 macrophages in alleviating the infammation of DN and explored the mechanism.Results: M2 macrophages hindered the progression of DN, reduced the levels of IL-1β (DN group was 34%, M2 group was 13%, p < 0.01) and MCP-1 (DN group was 49%, M2 group was 16%, p < 0.01) in the glomeruli. It was also proven that M2 macrophages alleviate mesangial cell injury caused by a high glucose environment. M2 macrophage tracking showed that the infused M2 macrophages migrated to the kidney, and the number of M2 macrophages in the kidney reached a maximum on day 3. Moreover, the ratio of M2 to M1 macrophages was 2.3 in the M2 infusion group, while 0.4 in the DN group (p < 0.01). Mechanistically, M2 macrophages downregulated Janus kinase (JAK) 2 and signal transducer and activator of transcription (STAT) 3 in mesangial cells.Conclusions: Multiple infusions of M2 macrophages significantly alleviated inflammation in the kidney and hindered the progression of DN at least partially by abrogating the M1/M2 homeostasis disturbances and suppressing the JAK2/STAT3 pathway in glomerular mesangial cells. M2 macrophage infusion may be a new therapeutic strategy for DN treatment.
Collapse
Affiliation(s)
- Yulin Gu
- Department of Endocrinology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Songyan Yu
- Department of Endocrinology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Weijun Gu
- Department of Endocrinology, Medical School of Chinese PLA, Chinese PLA General Hospital, Beijing, China
| | - Bing Li
- Department of Endocrinology, Medical School of Chinese PLA, Chinese PLA General Hospital, Beijing, China
| | - Jing Xue
- Department of Endocrinology, Medical School of Chinese PLA, Chinese PLA General Hospital, Beijing, China
| | - Jiejie Liu
- Department of Molecular Biology, Institute of Basic Medicine, School of Life Science, Chinese PLA General Hospital, Beijing, China
| | - Qi Zhang
- Department of Endocrinology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yaqi Yin
- Department of Endocrinology, Medical School of Chinese PLA, Chinese PLA General Hospital, Beijing, China
| | - Haixia Zhang
- Department of Endocrinology, Medical School of Chinese PLA, Chinese PLA General Hospital, Beijing, China
| | - Qinghua Guo
- Department of Endocrinology, Medical School of Chinese PLA, Chinese PLA General Hospital, Beijing, China
| | - Mingxia Yuan
- Department of Endocrinology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Zhaohui Lyu
- Department of Endocrinology, Medical School of Chinese PLA, Chinese PLA General Hospital, Beijing, China
| | - Yiming Mu
- Department of Endocrinology, Medical School of Chinese PLA, Chinese PLA General Hospital, Beijing, China
| | - Yu Cheng
- Department of Endocrinology, Medical School of Chinese PLA, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
12
|
Wei J, Tian Y, Wei J, Guan M, Yu X, Xie J, Fan G. Bilirubin regulates cell death type by alleviating macrophage mitochondrial dysfunction caused by cigarette smoke extract. Redox Rep 2024; 29:2382946. [PMID: 39074442 PMCID: PMC11288206 DOI: 10.1080/13510002.2024.2382946] [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: 07/31/2024] Open
Abstract
OBJECTIVES To explore the effects and mechanisms of bilirubin on mitochondrial function and type of macrophage cell death after exposure to cigarette smoke extract (CSE). METHODS RAW264.7 macrophages were treated with different concentrations of CSE and bilirubin solutions and divided into four groups: control, CSE, bilirubin, and bilirubin + CSE groups. The necrotic and apoptotic states of the macrophages were determined using an Annexin V-fluorescein 5-isothiocyanate/propidium iodide (FITC/PI) staining kit. Cytoplasmic NOD-like receptor family, pyrin domain containing 3 (NLRP3) expression in macrophages was detected by immunofluorescence and the levels of IL-1β and IL-18 in the supernatants of culture medium were detected by enzyme linked immunosorbent assay (ELISA) test. A JC-1 mitochondrial membrane potential detection kit was used to assess mitochondrial membrane damage and the adenosine triphosphate (ATP) assay kit was used to determine intracellular ATP levels. After the macrophages were stained with reactive oxygen species (ROS) specific dye, 2',7'-Dichlorodihydrofluorescein diacetate (DCFH-DA), the fluorescence intensity and proportion of ROS-positive macrophages were measured using flow cytometry. RESULTS We observed that compared with those of 0 μM (control group), concentrations of 5, 10, or 20 μΜ bilirubin significantly decreased cell viability, which was increased by bilirubin exposure below 1 μM. The effect of CSE on macrophage viability was concentration- and time-dependent. Bilirubin of 0.2 μM could alleviate the inhibition of macrophage viability caused by 5% CSE. In addition, bilirubin intervention could reduce the occurrence of necrosis and pyroptosis to a certain extent. CONCLUSIONS CSE could cause mitochondrial dysfunction in macrophages, as demonstrated by a decrease in mitochondrial membrane potential and intracellular ATP levels and an increase in ROS production, while bilirubin could relieve mitochondrial dysfunction caused by CSE.
Collapse
Affiliation(s)
- Jingjing Wei
- Department of Pediatrics, Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
| | - Yuan Tian
- Department of Pediatrics, Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
| | - Jinshu Wei
- Department of Pediatrics, Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
| | - Meiqi Guan
- Department of Pediatrics, Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
| | - Xiaoya Yu
- Department of Pediatrics, Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
| | - Jianing Xie
- Department of Pediatrics, Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
| | - Guoquan Fan
- School of Basic Medical Science, Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
| |
Collapse
|
13
|
Wang Y, Geng X, Guo Z, Chu D, Liu R, Cheng B, Cui H, Li C, Li J, Li Z. M2 macrophages promote subconjunctival fibrosis through YAP/TAZ signalling. Ann Med 2024; 56:2313680. [PMID: 38335557 PMCID: PMC10860428 DOI: 10.1080/07853890.2024.2313680] [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: 08/21/2023] [Accepted: 01/24/2024] [Indexed: 02/12/2024] Open
Abstract
PURPOSE To evaluate the role of M2 macrophages in subconjunctival fibrosis after silicone implantation (SI) and investigate the underlying mechanisms. MATERIALS AND METHODS A model of subconjunctival fibrosis was established by SI surgery in rabbit eyes. M2 distribution and collagen deposition were evaluated by histopathology. The effects of M2 cells on the migration (using wound-scratch assay) and activation (by immunofluorescence and western blotting) of human Tenon's fibroblasts (HTFs) were investigated. RESULTS There were more M2 macrophages (CD68+/CD206+ cells) occurring in tissue samples around silicone implant at 2 weeks postoperatively. Dense collagen deposition was observed at 8 weeks after SI. In vitro experiment showed M2 expressed high level of CD206 and transforming growth factor-β1 (TGF-β1). The M2-conditioned medium promoted HTFs migration and the synthesis of collagen I and fibronectin. Meanwhile, M2-conditioned medium increased the protein levels of TGF-β1, TGF-βR II, p-Smad2/3, yes-associated protein (YAP), and transcriptional coactivator with PDZ-binding motif (TAZ). Verteporfin, a YAP inhibitor, suppressedTGF-β1/Smad2/3-YAP/TAZ pathway and attenuated M2-induced extracellular matrix deposition by HTFs. CONCLUSIONS TGF-β1/Smad2/3-YAP/TAZ signalling may be involved in M2-induced fibrotic activities in HTFs. M2 plays a key role in promoting subconjunctival fibrosis and can serve as an attractive target for anti-fibrotic therapeutics.
Collapse
Affiliation(s)
- Yiwei Wang
- Henan Eye Hospital, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou, China
| | - Xingchen Geng
- Henan Eye Hospital, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhihua Guo
- Henan Eye Hospital, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou, China
| | - Dandan Chu
- Henan Eye Hospital, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou, China
| | - Ruixing Liu
- Henan Eye Hospital, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou, China
| | - Boyuan Cheng
- Henan Eye Hospital, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou, China
| | - Haohao Cui
- Henan Eye Hospital, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou, China
| | - Chengcheng Li
- Henan Eye Hospital, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou, China
| | - Jingguo Li
- Henan Eye Hospital, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhanrong Li
- Henan Eye Hospital, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
14
|
Zhang Q, Zhao L, Li Y, Wang S, Lu G, Wang H. Advances in the mechanism of action of short-chain fatty acids in psoriasis. Int Immunopharmacol 2024; 141:112928. [PMID: 39159566 DOI: 10.1016/j.intimp.2024.112928] [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: 05/31/2024] [Revised: 07/22/2024] [Accepted: 08/10/2024] [Indexed: 08/21/2024]
Abstract
Psoriasis is a prevalent chronic inflammatory and immunological disorder. Its lesions are present as scaly erythema or plaques. Disruptions in the body's immune system play a significant role in developing psoriasis. Recent evidence suggests a potential role of the gut microbiome in autoimmune diseases. Short-chain fatty acids (SCFAs) are the primary metabolites created by gut microbes and play a crucial fuction in autoimmunity. SCFAs act on various cells by mediating signaling to participate in host physiological and pathological processes. These processes encompass body metabolism, maintenance of intestinal barrier function, and immune system modulation. SCFAs can regulate immune cells to enhance the body's immune function, potentially influencing the prevention and treatment of psoriasis. However, the mechanisms underlying the role of SCFAs in psoriasis remain incompletely understood. This paper examines the relationship between SCFAs and psoriasis, elucidating how SCFAs influence the immune system, inflammatory response, and gut barrier in psoriasis. According to the study, in psoriasis, SCFAs have been shown to regulate neutrophils, macrophages, and dendritic cells in the adaptive immune system, as well as T and B cells in the innate immune system. Additionally, we explore the role of SCFAs in psoriasis by maintaining intestinal barrier function, restoring intestinal ecological homeostasis, and investigating the potential therapeutic benefits of SCFAs for psoriasis.
Collapse
Affiliation(s)
- Qin Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin, China; Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Linna Zhao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin, China; Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, China.
| | - Yu Li
- Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin, China; Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Siyao Wang
- Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin, China; Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Guiling Lu
- Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin, China
| | - Hongmei Wang
- Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin, China.
| |
Collapse
|
15
|
Dalpati N, Rai SK, Dash SP, Kumar P, Singh D, Sarangi PP. Integrins α5β1 and αvβ3 Differentially Participate in the Recruitment and Reprogramming of Tumor-associated Macrophages in the In Vitro and In Vivo Models of Breast Tumor. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:1553-1568. [PMID: 39330703 DOI: 10.4049/jimmunol.2400180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 09/10/2024] [Indexed: 09/28/2024]
Abstract
Tumor-associated macrophages (TAMs) drive the protumorigenic responses and facilitate tumor progression via matrix remodeling, angiogenesis, and immunosuppression by interacting with extracellular matrix proteins via integrins. However, the expression dynamics of integrin and its correlation with TAM functional programming in the tumors remain unexplored. In this study, we examined surface integrins' role in TAM recruitment and phenotypic programming in a 4T1-induced murine breast tumor model. Our findings show that integrin α5β1 is upregulated in CD11b+Ly6Chi monocytes in the bone marrow and blood by day 10 after tumor induction. Subsequent analysis revealed elevated integrin α5β1 expression on tumor-infiltrating monocytes (Ly6ChiMHC class II [MHCII]low) and M1 TAMs (F4/80+Ly6ClowMHCIIhi), whereas integrin αvβ3 was predominantly expressed on M2 TAMs (F4/80+Ly6ClowMHCIIlow), correlating with higher CD206 and MERTK expression. Gene profiling of cells sorted from murine tumors showed that CD11b+Ly6G-F4/80+α5+ TAMs had elevated inflammatory genes (IL-6, TNF-α, and STAT1/2), whereas CD11b+Ly6G-F4/80+αv+ TAMs exhibited a protumorigenic phenotype (IL-10, Arg1, TGF-β, and STAT3/6). In vitro studies demonstrated that blocking integrin α5 and αv during macrophage differentiation from human peripheral blood monocytes reduced cell spreading and expression of CD206 and CD163 in the presence of specific matrix proteins, fibronectin, and vitronectin. Furthermore, RNA sequencing data analysis (GEO dataset: GSE195857) from bone marrow-derived monocytes and TAMs in 4T1 mammary tumors revealed differential integrin α5 and αv expression and their association with FAK and SRC kinase. In line with this, FAK inhibition during TAM polarization reduced SRC, STAT1, and STAT6 phosphorylation. In conclusion, these findings underscore the crucial role of integrins in TAM recruitment, polarization, and reprogramming in tumors.
Collapse
Affiliation(s)
- Nibedita Dalpati
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, India
| | - Shubham Kumar Rai
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, India
| | - Shiba Prasad Dash
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, India
| | - Puneet Kumar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, India
| | - Divya Singh
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, India
| | - Pranita P Sarangi
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, India
| |
Collapse
|
16
|
Luo Y, Jiang LY, Liao ZZ, Wang YY, Wang YD, Xiao XH. Metabolic Regulation of Inflammation: Exploring the Potential Benefits of Itaconate in Autoimmune Disorders. Immunology 2024. [PMID: 39542834 DOI: 10.1111/imm.13875] [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: 09/08/2024] [Revised: 10/21/2024] [Accepted: 10/22/2024] [Indexed: 11/17/2024] Open
Abstract
Itaconic acid and its metabolites have demonstrated significant therapeutic potential in various immune diseases. Originating from the tricarboxylic acid cycle in immune cells, itaconic acid can modulate immune responses, diminish inflammation, and combat oxidative stress. Recent research has uncovered multiple mechanisms through which itaconic acid exerts its effects, including the inhibition of inflammatory cytokine production, activation of anti-inflammatory pathways, and modulation of immune cell function by regulating cellular metabolism. Cellular actions are influenced by the modulation of metabolic pathways, such as inhibiting succinate dehydrogenase (SDH) activity or glycolysis, activation of nuclear-factor-E2-related factor 2 (Nrf2), boosting cellular defences against oxidative stress, and suppression of immune cell inflammation through the NF-κB pathway. This comprehensive review discusses the initiation, progression, and mechanisms of action of itaconic acid and its metabolites, highlighting their modulatory effects on various immune cell types. Additionally, it examines their involvement in immune disease like rheumatoid arthritis, multiple sclerosis, type 1 diabetes mellitus, and autoimmune hepatitis, offering greater understanding for creating new therapies for these ailments.
Collapse
Affiliation(s)
- Yin Luo
- The First Affiliated Hospital, Department of Metabolism and Endocrinology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Li-Yan Jiang
- The First Affiliated Hospital, Department of Metabolism and Endocrinology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Zhe-Zhen Liao
- The First Affiliated Hospital, Department of Metabolism and Endocrinology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Yuan-Yuan Wang
- The First Affiliated Hospital, Department of Metabolism and Endocrinology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Ya-Di Wang
- The First Affiliated Hospital, Department of Metabolism and Endocrinology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Xin-Hua Xiao
- The First Affiliated Hospital, Department of Metabolism and Endocrinology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| |
Collapse
|
17
|
Feng N, Li Y, Guo F, Song J, Wang L, Li M, Gao K, Wang X, Chu D, Song Y, Wang L. Fibroblast growth factor 10 alleviates LPS-induced acute lung injury by promoting recruited macrophage M2 polarization. Inflammation 2024:10.1007/s10753-024-02158-4. [PMID: 39538090 DOI: 10.1007/s10753-024-02158-4] [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: 02/20/2024] [Revised: 07/03/2024] [Accepted: 09/30/2024] [Indexed: 11/16/2024]
Abstract
Acute lung injury (ALI) is characterized by damage to the alveoli and an overabundance of inflammation. Representing a serious inflammatory condition, ALI lacks a precise treatment approach. Despite the recognized benefit impacts of Fibroblast growth factor-10 (FGF10) on ALI, the underlying mechanisms remain unelucidated. To study the role of FGF10 in ALI, C57BL/6 J mice were intratracheally injected with 5 mg/kg Lipopolysaccharide (LPS) with FGF10 (5 mg/kg) or an equal volume of PBS. Inflammatory factors were quantified in bronchoalveolar lavage fluid (BALF) and plasma using ELISA. RNA sequencing of F4/80+Ly6G- macrophages in BALF explored changes in macrophage phenotype and potential mechanisms. Macrophage polarization in BALF was assessed using qRT-PCR, flow cytometry, and Western blot analysis. In vitro, a Transwell co-culture of mouse lung epithelial cells (MLE12) and bone marrow macrophages (BMDM) validated the role of FGF10 in modulating LPS-induced macrophage phenotypic changes. FGF10 ameliorated LPS-induced ALI by diminishing pro-inflammatory factors (IL-1β, TNF-α, and IL-6) and the neutrophil accumulation in BALF. FGF10 also increased the levels of anti-inflammatory factor IL-10. The FGF10 intervention group exhibited enhanced gene expression of macrophage arginine biosynthesis marker (ARG1), and expression of M2-type marker CD206 in monocytes and macrophages. In addition, phosphorylated STAT3 expression increased in isolated monocyte-derived macrophages. Experiments in vitro confirmed that FGF10 could elevate macrophage M2 marker ARG1 expression through the JAK2/STAT3 pathway. FGF10 ameliorates acute LPS-induced lung injury by modulating the polarization of monocyte-derived macrophages recruited in the alveolar space to the M2 type.
Collapse
Affiliation(s)
- Nana Feng
- Department of Respiratory and Critical Medicine, Shanghai Eighth People's Hospital, Shanghai, 200235, China
| | - Yufan Li
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Fengxia Guo
- Department of Respiratory and Critical Medicine, Shanghai Eighth People's Hospital, Shanghai, 200235, China
| | - Juan Song
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Lu Wang
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Miao Li
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Kaijing Gao
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Xiaocen Wang
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Dejie Chu
- Department of Respiratory and Critical Medicine, Shanghai Eighth People's Hospital, Shanghai, 200235, China.
| | - Yuanlin Song
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
- Shanghai Key Laboratory of Lung Inflammation and Injury, Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
- Shanghai Institute of Infectious Disease and Biosecurity, Shanghai, 200032, China.
- Shanghai Respiratory Research Institute, Shanghai, 200032, China.
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, 200032, China.
- Jinshan Hospital of Fudan University, Shanghai, 201508, China.
| | - Linlin Wang
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
18
|
Xin L, Liu J, Lai JY, Xu HS, Fan LJ, Zou YH, Zhou Q, Yue ZQ, Gan JH. Methionine restriction promotes the polarization of macrophages towards M1 and the immunotherapy effect of PD-L1/PD-1 blockades by inhibiting the secretion of MIF by gastric carcinoma cells. Transl Oncol 2024; 51:102181. [PMID: 39541710 DOI: 10.1016/j.tranon.2024.102181] [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: 04/16/2024] [Revised: 09/13/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND The limited curative effect of PD-L1/PD-1 blockades presents challenges to immunotherapy for advanced gastric cancer. We have found that methionine restriction (MR) enhances the drug resistance of gastric carcinoma cells. We aimed to explore whether MR can enhance the efficacy of PD-L1/PD-1 blockades in gastric cancer. METHODS To conduct MR, gastric carcinoma cells were transfected with LV-METase in vitro, and 615 mice were injected with MFC cells with stable METase expression in vivo. Flow cytometry was conducted to measure the proportions of M1/M2 macrophages and CD8+ GZMB+/IFN-γ+ T cells. Additionally, the levels of M1/M2 macrophage markers and MIF were also detected. RESULTS MR increased M1 and down-regulated M2 macrophages. MR suppressed MIF levels in gastric carcinoma cells, while the addition of anti-MIF neutralizing antibody inhibited the effect of MR on macrophage M1/M2 polarization. MR enhanced the increase of the proportion of CD8+ GZMB+ T cells and CD8+ IFN-γ+ T cells induced by PD-L1/PD-1 blockades. In vivo detection verified the efficacy of the combination of MR and PD-L1/PD-1 blockades on gastric cancer. CONCLUSIONS MR inhibits the secretion of MIF by gastric carcinoma cells, promotes macrophage M1 polarization, and enhances the therapeutic effect of PD-L1/PD-1 blockades in gastric cancer.
Collapse
Affiliation(s)
- Lin Xin
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi Province, China; Intelligent Medical Imaging of Jiangxi Key Laboratory, Nanchang 330006, China.
| | - Jiang Liu
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi Province, China
| | - Jun-Yan Lai
- Class 2210, The Second clinical medical college, Nanchang University, Nanchang, 330006, Jiangxi Province, China
| | - He-Song Xu
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi Province, China
| | - Luo-Jun Fan
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi Province, China
| | - Yong-Hui Zou
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi Province, China
| | - Qi Zhou
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi Province, China
| | - Zhen-Qi Yue
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi Province, China
| | - Jin-Heng Gan
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi Province, China
| |
Collapse
|
19
|
Chen B, Zhao X, Xu M, Luo J, Bai L, Han Q, Gao Y, Guo B, Yin Z. Inflammation-Responsive Functional Core-Shell Micro-Hydrogels Promote Rotator Cuff Tendon-To-Bone Healing by Recruiting MSCs and Immuno-Modulating Macrophages in Rats. Adv Healthc Mater 2024:e2404091. [PMID: 39526494 DOI: 10.1002/adhm.202404091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Indexed: 11/16/2024]
Abstract
Rotator cuff injuries often necessitate surgical intervention, but the outcomes are often unsatisfactory. The underlying reasons can be attributed to multiple factors, with the intricate inflammatory activities and insufficient presence of stem cells being particularly significant. In this study, an innovative inflammation-responsive core-shell micro-hydrogel is designed for independent release of SDF-1 and IL-4 within a single delivery system to promote tendon-to-bone healing by recruiting MSCs and modulating M2 macrophages polarization. First, a MMP-2 responsive hydrogel loaded with IL-4 (GelMA-MMP/IL-4) is synthesized by cross-linking gelatin methacrylate (GelMA) with MMP-2 substrate peptide. Then, the resulting core particles are coated with a shell of chitosan /SDF-1/hyaluronic acid (CS/HA/SDF-1) using the layer-by-layer electrostatic deposition method to form a core-shell micro-hydrogel composite. The core-shell micro-hydrogel shows sustained release of SDF-1 and MMP-2-responsive release of IL-4 associated in situ MSCs homing and smart inflammation regulation by promoting M2 macrophages polarization. Additionally, by injecting these micro-hydrogels into a rat rotator cuff tear and repair model, notable improvements of fibrocartilage layer are observed between tendon and bone. Notably, this study presents a new and potentially powerful environment-responsive drug delivery strategy that offers valuable insights for regulating the intricate micro-environment associated with tissue regeneration.
Collapse
Affiliation(s)
- Baojun Chen
- Department of Surgery of Spine and Spinal Cord, Henan Provincial People's Hospital, Henan Province Intelligent orthopedic technology innovation and transformation International Joint Laboratory, Henan Key Laboratory for intelligent precision orthopedics, People's Hospital of Zhengzhou University, Zhengzhou, 450003, China
| | - Xin Zhao
- Frontier Institute of Science and Technology, and State Key Laboratory for Mechanical Behavior of Materials, and Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Meiguang Xu
- Department of Orthopaedics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Jinlong Luo
- Frontier Institute of Science and Technology, and State Key Laboratory for Mechanical Behavior of Materials, and Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Lang Bai
- Department of Orthopaedics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Qian Han
- Department of Orthopaedics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Yanzheng Gao
- Department of Surgery of Spine and Spinal Cord, Henan Provincial People's Hospital, Henan Province Intelligent orthopedic technology innovation and transformation International Joint Laboratory, Henan Key Laboratory for intelligent precision orthopedics, People's Hospital of Zhengzhou University, Zhengzhou, 450003, China
| | - Baolin Guo
- Frontier Institute of Science and Technology, and State Key Laboratory for Mechanical Behavior of Materials, and Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Zhanhai Yin
- Department of Orthopaedics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| |
Collapse
|
20
|
Yorek M, Jiang X, Liu S, Hao J, Yu J, Avellino A, Liu Z, Curry M, Keen H, Shao J, Kanagasabapathy A, Kong M, Xiong Y, Sauter ER, Sugg SL, Li B. FABP4-mediated lipid accumulation and lipolysis in tumor-associated macrophages promote breast cancer metastasis. eLife 2024; 13:RP101221. [PMID: 39513934 PMCID: PMC11548877 DOI: 10.7554/elife.101221] [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] [Indexed: 11/16/2024] Open
Abstract
A high density of tumor-associated macrophages (TAMs) is associated with poorer prognosis and survival in breast cancer patients. Recent studies have shown that lipid accumulation in TAMs can promote tumor growth and metastasis in various models. However, the specific molecular mechanisms that drive lipid accumulation and tumor progression in TAMs remain largely unknown. Herein, we demonstrated that unsaturated fatty acids (FAs), unlike saturated ones, are more likely to form lipid droplets in murine macrophages. Specifically, unsaturated FAs, including linoleic acids (LA), activate the FABP4/CEBPα pathway, leading to triglyceride synthesis and lipid droplet formation. Furthermore, FABP4 enhances lipolysis and FA utilization by breast cancer cell lines, which promotes cancer cell migration in vitro and metastasis in vivo. Notably, a deficiency of FABP4 in murine macrophages significantly reduces LA-induced lipid metabolism. Therefore, our findings suggest FABP4 as a crucial lipid messenger that facilitates unsaturated FA-mediated lipid accumulation and lipolysis in TAMs, thus contributing to the metastasis of breast cancer.
Collapse
Affiliation(s)
- Matthew Yorek
- Department of Pathology, Holden Comprehensive Cancer Center, University of IowaIowa CityUnited States
| | - Xingshan Jiang
- Department of Pathology, Holden Comprehensive Cancer Center, University of IowaIowa CityUnited States
| | - Shanshan Liu
- Department of Pathology, Holden Comprehensive Cancer Center, University of IowaIowa CityUnited States
| | - Jiaqing Hao
- Department of Pathology, Holden Comprehensive Cancer Center, University of IowaIowa CityUnited States
| | - Jianyu Yu
- Department of Pathology, Holden Comprehensive Cancer Center, University of IowaIowa CityUnited States
| | - Anthony Avellino
- Department of Pathology, Holden Comprehensive Cancer Center, University of IowaIowa CityUnited States
| | - Zhanxu Liu
- Department of Bioinformatics and Biostatistics, University of LouisvilleLouisvilleUnited States
| | - Melissa Curry
- Holden Comprehensive Cancer Center, University of Iowa Hospitals and ClinicsIowa CityUnited States
| | - Henry Keen
- Iowa Institute of Human Genetics, University of IowaIowa CityUnited States
| | - Jianqiang Shao
- Central Microscopy Research Facility, University of IowaIowa CityUnited States
| | - Anand Kanagasabapathy
- Department of Pathology, Holden Comprehensive Cancer Center, University of IowaIowa CityUnited States
| | - Maying Kong
- Department of Bioinformatics and Biostatistics, University of LouisvilleLouisvilleUnited States
| | - Yiqin Xiong
- Department of Pathology, Holden Comprehensive Cancer Center, University of IowaIowa CityUnited States
| | - Edward R Sauter
- Division of Cancer Prevention, NIH/NCIRockvilleUnited States
| | - Sonia L Sugg
- Department of Surgery, University of IowaIowa CityUnited States
| | - Bing Li
- Department of Pathology, Holden Comprehensive Cancer Center, University of IowaIowa CityUnited States
| |
Collapse
|
21
|
Zhang A, Jiang J, Zhang C, Xu H, Yu W, Zhang ZN, Yuan L, Lu Z, Deng Y, Fan H, Fang C, Wang X, Shao A, Chen S, Li H, Ni J, Wang W, Zhang X, Zhang J, Luan B. Thermogenic Adipocytes Promote M2 Macrophage Polarization through CNNM4-Mediated Mg Secretion. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024:e2401140. [PMID: 39517124 DOI: 10.1002/advs.202401140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 10/09/2024] [Indexed: 11/16/2024]
Abstract
M2 macrophages promote adipose tissue thermogenesis which dissipates energy in the form of heat to combat obesity. However, the regulation of M2 macrophages by thermogenic adipocytes is unclear. Here, it is identified magnesium (Mg) as a thermogenic adipocyte-secreted factor to promote M2 macrophage polarization. Mg transporter Cyclin and CBS domain divalent metal cation transport mediator 4 (CNNM4) induced by ADRB3-PKA-CREB signaling in thermogenic adipocytes during cold exposure mediates Mg efflux and Mg in turn binds to the DFG motif in mTOR to facilitate mTORC2 activation and M2 polarization in macrophages. In obesity, downregulation of CNNM4 expression inhibits Mg secretion from thermogenic adipocytes, which leads to decreased M2 macrophage polarization and thermogenesis. As a result, CNNM4 overexpression in adipocytes or Mg supplementation in adipose tissue ameliorates obesity by promoting thermogenesis. Importantly, an Mg wire implantation (AMI) approach is introduced to achieve adipose tissue-specific long-term Mg supplement. AMI promotes M2 macrophage polarization and thermogenesis and ameliorates obesity in mice. Taken together, a reciprocal regulation of thermogenic adipocytes and M2 macrophages important for thermogenesis is identified, and AMI is offered as a promising strategy against obesity.
Collapse
Affiliation(s)
- Anke Zhang
- Department of Endocrinology, Tongji Hospital Affiliated to Tongji University, School of Medicine, Tongji University, Shanghai, 200065, P. R. China
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, P. R. China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, 310009, P. R. China
| | - Junkun Jiang
- Department of Endocrinology, Tongji Hospital Affiliated to Tongji University, School of Medicine, Tongji University, Shanghai, 200065, P. R. China
| | - Chuan Zhang
- Department of Endocrinology, Tongji Hospital Affiliated to Tongji University, School of Medicine, Tongji University, Shanghai, 200065, P. R. China
| | - Houshi Xu
- Department of Neurosurgery, Huashan Hospital Affiliated to Fudan University, School of Medicine, Fudan University, Shanghai, 200040, P. R. China
| | - Wenjing Yu
- Department of Endocrinology, Tongji Hospital Affiliated to Tongji University, School of Medicine, Tongji University, Shanghai, 200065, P. R. China
| | - Zhen-Ning Zhang
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200120, China
| | - Ling Yuan
- School of Public Health, School of Medicine, Shanghai Jiaotong University, Shanghai, 200025, China
| | - Zhangming Lu
- Department of Endocrinology, Tongji Hospital Affiliated to Tongji University, School of Medicine, Tongji University, Shanghai, 200065, P. R. China
| | - Yuqing Deng
- Department of Endocrinology, Tongji Hospital Affiliated to Tongji University, School of Medicine, Tongji University, Shanghai, 200065, P. R. China
| | - Haonan Fan
- Department of Endocrinology, Tongji Hospital Affiliated to Tongji University, School of Medicine, Tongji University, Shanghai, 200065, P. R. China
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, P. R. China
| | - Chaoyou Fang
- Department of Neurosurgery, Shanghai General Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200080, P. R. China
| | - Xiaoyu Wang
- Department of Endocrinology, Tongji Hospital Affiliated to Tongji University, School of Medicine, Tongji University, Shanghai, 200065, P. R. China
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, P. R. China
| | - Anwen Shao
- Department of Endocrinology, Tongji Hospital Affiliated to Tongji University, School of Medicine, Tongji University, Shanghai, 200065, P. R. China
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, P. R. China
| | - Sheng Chen
- Department of Endocrinology, Tongji Hospital Affiliated to Tongji University, School of Medicine, Tongji University, Shanghai, 200065, P. R. China
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, P. R. China
| | - Huaming Li
- Department of Endocrinology, Tongji Hospital Affiliated to Tongji University, School of Medicine, Tongji University, Shanghai, 200065, P. R. China
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, P. R. China
| | - Jiahua Ni
- College of Biological Science and Medical Engineering, Donghua University, 2999 North Renmin Road, Shanghai, 201620, China
| | - Wenhui Wang
- College of Biological Science and Medical Engineering, Donghua University, 2999 North Renmin Road, Shanghai, 201620, China
| | - Xiaonong Zhang
- State Key Laboratory of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
- Suzhou Origin Medical Technology Co. Ltd., Suzhou, 215513, China
| | - Jianmin Zhang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, P. R. China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, 310009, P. R. China
- Brain Research Institute, Zhejiang University, Hangzhou, 310009, P. R. China
- MOE Frontier Science Center for Brain Science and Brain-Machine Integration, Zhejiang University, Hangzhou, 310009, P. R. China
| | - Bing Luan
- Department of Endocrinology, Tongji Hospital Affiliated to Tongji University, School of Medicine, Tongji University, Shanghai, 200065, P. R. China
| |
Collapse
|
22
|
Yang L, Ren Q, Wang Y, Zheng Y, Du F, Wang F, Zhou J, Gui L, Chen S, Chen X, Zhang W, Sun Y, Zhong X, Liu H, Jiang X, Zhang Z. Research progress of mitochondrial dysfunction induced pyroptosis in acute lung injury. Respir Res 2024; 25:398. [PMID: 39511593 PMCID: PMC11545853 DOI: 10.1186/s12931-024-03028-1] [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: 04/01/2024] [Accepted: 10/25/2024] [Indexed: 11/15/2024] Open
Abstract
Acute lung injury (ALI) is a common critical respiratory disease in clinical practice, especially in the ICU, with a high mortality rate. The pathogenesis of ALI is relatively complex, mainly involving inflammatory response imbalance, oxidative stress, cell apoptosis, and other aspects. However, currently, the treatment measures taken based on the above mechanisms have not had significant effects. Recent research shows that mitochondrial dysfunction and pyroptosis play an important role in ALI, but there is not much analysis on the relationship between mitochondrial dysfunction and pyroptosis at present. This article reviews the situation of mitochondrial dysfunction in ALI, pyroptosis in ALI, whether mitochondrial dysfunction is related to pyroptosis in ALI, and how to do so, and further analyzes the relationship between them in ALI. This review describes how to alleviate mitochondrial dysfunction, and then suppress the associated immunological pyroptosis, providing new ideas for the clinical treatment of ALI.
Collapse
Affiliation(s)
- Luhan Yang
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Qingyi Ren
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Yaohui Wang
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Yucheng Zheng
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Fei Du
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Fang Wang
- Department of Gastroenterology, Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Jie Zhou
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Linxi Gui
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Shengdong Chen
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Xiurui Chen
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Wanting Zhang
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Yuhong Sun
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Xiaolin Zhong
- Department of Gastroenterology, Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Hao Liu
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China.
| | - Xian Jiang
- Department of Anesthesiology, Luzhou People's Hospital, Luzhou, 646000, China.
| | - Zhuo Zhang
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China.
| |
Collapse
|
23
|
Smyth T, Payton A, Hickman E, Rager JE, Jaspers I. Leveraging a comprehensive unbiased RNAseq database to characterize human monocyte-derived macrophage gene expression profiles within commonly employed in vitro polarization methods. Sci Rep 2024; 14:26753. [PMID: 39500943 PMCID: PMC11538326 DOI: 10.1038/s41598-024-78000-6] [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: 04/05/2024] [Accepted: 10/28/2024] [Indexed: 11/08/2024] Open
Abstract
Macrophages are pivotal innate immune cells which exhibit high phenotypic plasticity and can exist in different polarization states dependent on exposure to external stimuli. Numerous methods have been employed to simulate macrophage polarization states to test their function in vitro. However, limited research has explored whether these polarization methods yield comparable populations beyond key gene, cytokine, and cell surface marker expression. Here, we employ an unbiased comprehensive analysis using data organized through the all RNA-seq and ChIP-seq sample and signature search (ARCHS4) database, which compiles all RNAseq data deposited into the National Center for Biotechnology Information (NCBI) Sequence Read Archive (SRA). In silico analyses were carried out demonstrating that commonly employed macrophage polarization methods generate distinct gene expression profiles in macrophage subsets that remained poorly described until now. Our analyses confirm existing knowledge on broad macrophage polarization, while expanding nuanced differences between M2a and M2c subsets, suggesting non-interchangeable stimuli for M2a polarization. Furthermore, we characterize divergent gene expression patterns in M1 macrophages following standard polarization protocols, indicating significant subset distinctions. Consequently, equivalence cannot be assumed among polarization regimens for in vitro macrophage studies, particularly in simulating diverse pathogen responses.
Collapse
Affiliation(s)
- Timothy Smyth
- Curriculum in Toxicology & Environmental Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Center for Environmental Medicine, Asthma, and Lung Biology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Alexis Payton
- Center for Environmental Medicine, Asthma, and Lung Biology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Elise Hickman
- Curriculum in Toxicology & Environmental Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Environmental Sciences and Engineering, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Julia E Rager
- Curriculum in Toxicology & Environmental Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Center for Environmental Medicine, Asthma, and Lung Biology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Environmental Sciences and Engineering, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Ilona Jaspers
- Curriculum in Toxicology & Environmental Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Center for Environmental Medicine, Asthma, and Lung Biology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Department of Pediatrics, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- , 116 Manning Drive, Campus Box 7310, Chapel Hill, NC, 27599-7310, USA.
| |
Collapse
|
24
|
Sun X, Zhang J, Dong B, Xiong Q, Wang X, Gu Y, Wang Z, Liu H, Zhang J, He X, Liu H, Zhong Y, Yi C, Chi X, Liu Z, Pang X, Cui Y. Targeting SLITRK4 Restrains Proliferation and Liver Metastasis in Colorectal Cancer via Regulating PI3K/AKT/NFκB Pathway and Tumor-Associated Macrophage. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024:e2400367. [PMID: 39499724 DOI: 10.1002/advs.202400367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 10/28/2024] [Indexed: 11/07/2024]
Abstract
Liver metastasis is the major cause of death in colorectal cancer (CRC) due to the lack of effective treatment. To explore novel drivers of CRC liver metastasis, the transcriptomes of primary paracancerous, colorectal tumors and metastases from human patients are profiled. It is found that SLIT- and NTRK-like family member 4 (SLITRK4) is the top upregulated gene in liver metastases and is associated with worse overall survival of CRC patients. Multiple in vitro and in vivo models suggested SLITRK4 promoted CRC tumorigenesis, invasion, migration, and angiogenesis, and inhibition of it restrained CRC tumor growth and liver metastasis with a more profound effect on the tumor microenvironment (TME). Mechanistically, SLITRK4 overexpression significantly activated the PI3K/AKT/NFκB pathway, regulated extracellular matrix organization, and multiple cytokines expression. Furthermore, the results from coculture models and single-cell RNA sequencing analyses suggested SLITRK4 promoted tumor-associated macrophages (TAMs) infiltration and polarization. In addition, macrophage depletion significantly inhibited SLITRK4-induced liver metastasis in CRC. Finally, pharmacological inhibition of SLITRK4 by using lipid-polymer hybrid nanoparticles (NPs) for systemic siRNA delivery can effectively inhibit CRC liver metastasis. Taken together, these results pinpoint that SLITRK4 regulates CRC tumorigenesis and liver metastasis, and siRNA delivering NPs agents validate the therapeutic potential of targeting SLITRK4 in CRC.
Collapse
Affiliation(s)
- Xiaojiao Sun
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Junling Zhang
- Department of General Surgery, Peking University First Hospital, Xishiku Street, Beijing, Xicheng, 100034, China
| | - Bingqi Dong
- Department of General Surgery, Peking University First Hospital, Xishiku Street, Beijing, Xicheng, 100034, China
| | - Qingqing Xiong
- Department of Hepatobiliary Cancer, Liver Cancer Center, Tianjin Medical University Cancer Institute, Tianjin, 300060, China
| | - Xin Wang
- Department of General Surgery, Peking University First Hospital, Xishiku Street, Beijing, Xicheng, 100034, China
| | - Yanlun Gu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
- Department of Pharmacy, Peking University First Hospital, Xishiku Street, Beijing, Xicheng, 100034, China
- Institute of Clinical Pharmacology, Peking University, Xueyuan Road 38, Beijing, Haidian, 100191, China
| | - Zhiqi Wang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Huiyu Liu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Jixin Zhang
- Department of Pathology, Peking University First Hospital, Xishiku Street, Beijing, Xicheng, 100034, China
| | - Xu He
- Department of Pharmacy, Peking University First Hospital, Xishiku Street, Beijing, Xicheng, 100034, China
- Institute of Clinical Pharmacology, Peking University, Xueyuan Road 38, Beijing, Haidian, 100191, China
| | - Hongjin Liu
- Department of General Surgery, Peking University First Hospital, Xishiku Street, Beijing, Xicheng, 100034, China
| | - Yi Zhong
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Chuxiao Yi
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Xiaowei Chi
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Zhenming Liu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Xiaocong Pang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
- Department of Pharmacy, Peking University First Hospital, Xishiku Street, Beijing, Xicheng, 100034, China
- Institute of Clinical Pharmacology, Peking University, Xueyuan Road 38, Beijing, Haidian, 100191, China
| | - Yimin Cui
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
- Department of Pharmacy, Peking University First Hospital, Xishiku Street, Beijing, Xicheng, 100034, China
- Institute of Clinical Pharmacology, Peking University, Xueyuan Road 38, Beijing, Haidian, 100191, China
| |
Collapse
|
25
|
Xiang X, Wang K, Zhang H, Mou H, Shi Z, Tao Y, Song H, Lian Z, Wang S, Lu D, Wei X, Xie H, Zheng S, Wang J, Xu X. Blocking CX3CR1+ Tumor-Associated Macrophages Enhances the Efficacy of Anti-PD1 Therapy in Hepatocellular Carcinoma. Cancer Immunol Res 2024; 12:1603-1620. [PMID: 39115356 DOI: 10.1158/2326-6066.cir-23-0627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 04/08/2024] [Accepted: 08/07/2024] [Indexed: 11/05/2024]
Abstract
The efficacy of immune checkpoint inhibitors in the treatment of hepatocellular carcinoma (HCC) remains limited, highlighting the need for further investigation into the mechanisms underlying treatment resistance. Accumulating evidence indicates that tumor-associated macrophages (TAM) within the tumor microenvironment demonstrate a key role in immune evasion and treatment resistance. This study explored the role of TAMs in the HCC tumor microenvironment. Our findings reveal that TAMs expressing CX3C motif chemokine receptor 1 (CX3CR1) induced T-cell exhaustion through IL27 secretion in orthotopic models of HCC following treatment with anti-PD1. Moreover, we identified prostaglandin E2 (PGE2), released by immune-attacked tumor cells, as a key regulator of TAM transition to a CX3CR1+ phenotype. To augment the therapeutic response to anti-PD1 therapy, we propose targeting CX3CR1+ TAMs in addition to anti-PD1 therapy. Our study contributes to the understanding of the role of TAMs in cancer immunotherapy and highlights potential clinical implications for HCC treatment. The combination of targeting CX3CR1+ TAMs with anti-PD1 therapy holds promise for enhancing the efficacy of immunotherapeutic interventions in patients with HCC.
Collapse
Affiliation(s)
- Xiaonan Xiang
- Zhejiang University School of Medicine, Hangzhou, China
| | - Kai Wang
- School of Clinical Medicine, Hangzhou Medical College, Hangzhou, China
| | - Hui Zhang
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Hangzhou, China
| | - Haibo Mou
- Department of Medical Oncology, Shulan (Hangzhou) Hospital, Hangzhou, China
| | - Zhixiong Shi
- Zhejiang University School of Medicine, Hangzhou, China
| | - Yaoye Tao
- Zhejiang University School of Medicine, Hangzhou, China
| | - Hongliang Song
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Hangzhou, China
| | - Zhengxing Lian
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Hangzhou, China
| | - Shuai Wang
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Hangzhou, China
| | - Di Lu
- School of Clinical Medicine, Hangzhou Medical College, Hangzhou, China
| | - Xuyong Wei
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Hangzhou, China
| | - Haiyang Xie
- Department of Hepatobiliary & Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, China
| | - Shusen Zheng
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, China
- Department of Hepatobiliary & Pancreatic Surgery, Shulan (Hangzhou) Hospital, Hangzhou, China
| | - Jianguo Wang
- Department of Hepatobiliary and Pancreatic Surgery, Minimally Invasive Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, China
| | - Xiao Xu
- School of Clinical Medicine, Hangzhou Medical College, Hangzhou, China
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
26
|
Jiang S, Cui Y, Wang B, Fu Z, Dong C. Acidic polysaccharides from Cistanche deserticola and their effects on the polarization of tumor-associated macrophages. Int J Biol Macromol 2024; 282:137207. [PMID: 39491707 DOI: 10.1016/j.ijbiomac.2024.137207] [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: 06/27/2024] [Revised: 10/08/2024] [Accepted: 10/31/2024] [Indexed: 11/05/2024]
Abstract
Three purified polysaccharides, CDAP-1, CDAP-2, and CDAP-3, were prepared from the rhizome of Cistanche deserticola and characterized. Structural analysis revealed that CDAP-1 and CDAP-2 are highly branched RG-I-type polysaccharides with side chains, including arabinans, galactans, and/or AGs, whereas CDAP-3 is a typical HG-type polysaccharide. In vivo tests revealed that treatment with the crude polysaccharide fraction (CDCP) significantly prolonged the survival of H22 tumor-bearing mice and exhibited antitumor effects. In vitro experiments demonstrated that all three polysaccharides could polarize M2-like TAMs toward the M1 phenotype. As a major component of CDCP, CDAP-2 could act on M2 macrophages through the TLR4 receptor-mediated NF-κB signaling pathway. An in vitro cell model verified that CDAP-2 could inhibit cell proliferation by reversing the polarization of M2-like TAMs to the cytotoxic M1 phenotype. Overall, we found that CDCP showed a clear antitumor effect and that its major component, CDAP-2, could reverse the suppressive TAM phenotype in the microenvironment, providing a scientific basis for the clinical application and development of C. deserticola.
Collapse
Affiliation(s)
- Siliang Jiang
- Tianjin Key Laboratory of Technologies Enabling Development of Clinical Therapeutics and Diagnosis, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China; Key Laboratory of Basic and Application Research of Beiyao, Heilongjiang University of Chinese Medicine, Ministry of Education, 24 Heping Road, Harbin 150040, China
| | - Yongsheng Cui
- Tianjin Key Laboratory of Technologies Enabling Development of Clinical Therapeutics and Diagnosis, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Bo Wang
- College of Pharmacy, Ningxia Medical University, No.692 Sheng-Li Street, Xing-Qing District, Yinchuan 750004, China
| | - Zheng Fu
- Department of Immunology, Key Laboratory of Immune Microenvironment and Disease of the Educational Ministry of China, Tianjin Key Laboratory of Cellular and Molecular Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China; Kangzhe Pharmaceutical Technology Development Company, Ltd., Tianjin, China
| | - Caixia Dong
- Tianjin Key Laboratory of Technologies Enabling Development of Clinical Therapeutics and Diagnosis, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China.
| |
Collapse
|
27
|
Yang X, Gao X, Xu C, Ni T, Sheng Y, Wang J, Sun X, Yuan J, Zhang L, Wang Y. Cryoablation synergizes with anti-PD-1 immunotherapy induces an effective abscopal effect in murine model of cervical cancer. Transl Oncol 2024; 51:102175. [PMID: 39489086 DOI: 10.1016/j.tranon.2024.102175] [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: 06/06/2024] [Revised: 09/15/2024] [Accepted: 10/29/2024] [Indexed: 11/05/2024] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs), especially anti-PD-1/PD-L1 antibodies, have emerged as promising therapeutic options for cervical cancer. However, the efficacy of anti-PD-1 antibody monotherapy is limited. Cryoablation could elicit an anti-tumor immune response, thereby presenting itself as a potential approach to augment the response of ICIs. The aim of our study was to investigate the systemic immunological effects of cryoablation and the potential synergistic anti-tumor effects of cryoablation and anti-PD-1 antibody in cervical cancer. METHODS We established U14 murine bilateral subcutaneous cervical cancer model, wherein the primary tumors were treated with cryoablation. Flow cytometry, immunohistochemistry and RNA-seq were used to analyze the immune cell infiltration and immune-associated pathways in the secondary tumor. RESULTS Our study revealed that cryoablation reprogrammed the immune landscape, leading to an enhanced infiltration of CD8+ T cell in distant tumors. Cryoablation created a conducive environment for increasing the efficacy of anti-PD-1 immunotherapy. Cryoablation in combination with anti-PD-1 antibody inhibited distant tumors growth and improved mouse survival. Mechanistically, this combination therapy could augment the infiltration of CD8+ T cells, CD4+ T cells, dendritic cells and M1-like tumor-associated macrophages, enhance multiple aspects of antitumor immune response, and reduce immunosuppressive cells such as M2-like tumor-associated macrophages and myeloid-derived suppressor cells in distant tumors. CONCLUSIONS Combination therapy with cryoablation and anti-PD-1 antibody induces an effective abscopal effect in murine model of cervical cancer and may be a novel therapeutic approach for patients with advanced/recurrent cervical cancer.
Collapse
Affiliation(s)
- Xiaoming Yang
- Department of Gynecologic Oncology, the International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China; Shanghai Municipal Key Clinical Specialty of gynecologic oncology, Shanghai 200030, China; Shanghai Key Laboratory of Embryo Original Diseases Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Xiaoyan Gao
- Department of Gynecologic Oncology, the International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China; Shanghai Municipal Key Clinical Specialty of gynecologic oncology, Shanghai 200030, China; Shanghai Key Laboratory of Embryo Original Diseases Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Chen Xu
- Department of Gynecologic Oncology, the International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China; Shanghai Municipal Key Clinical Specialty of gynecologic oncology, Shanghai 200030, China; Shanghai Key Laboratory of Embryo Original Diseases Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Ting Ni
- Department of Gynecologic Oncology, the International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China; Shanghai Municipal Key Clinical Specialty of gynecologic oncology, Shanghai 200030, China; Shanghai Key Laboratory of Embryo Original Diseases Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Yaru Sheng
- Department of Gynecologic Oncology, the International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China; Shanghai Municipal Key Clinical Specialty of gynecologic oncology, Shanghai 200030, China; Shanghai Key Laboratory of Embryo Original Diseases Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Jing Wang
- Department of Gynecologic Oncology, the International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China; Shanghai Municipal Key Clinical Specialty of gynecologic oncology, Shanghai 200030, China; Shanghai Key Laboratory of Embryo Original Diseases Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Xiao Sun
- Shanghai Municipal Key Clinical Specialty of gynecologic oncology, Shanghai 200030, China; Shanghai Key Laboratory of Embryo Original Diseases Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Jiangjing Yuan
- Department of Gynecologic Oncology, the International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China; Shanghai Municipal Key Clinical Specialty of gynecologic oncology, Shanghai 200030, China; Shanghai Key Laboratory of Embryo Original Diseases Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Lin Zhang
- Department of Gynecologic Oncology, the International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China; Shanghai Municipal Key Clinical Specialty of gynecologic oncology, Shanghai 200030, China; Shanghai Key Laboratory of Embryo Original Diseases Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China.
| | - Yudong Wang
- Department of Gynecologic Oncology, the International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China; Shanghai Municipal Key Clinical Specialty of gynecologic oncology, Shanghai 200030, China; Shanghai Key Laboratory of Embryo Original Diseases Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China.
| |
Collapse
|
28
|
Almansour S, Dunster JL, Crofts JJ, Nelson MR. Modelling the continuum of macrophage phenotypes and their role in inflammation. Math Biosci 2024; 377:109289. [PMID: 39243940 DOI: 10.1016/j.mbs.2024.109289] [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: 04/23/2024] [Revised: 07/15/2024] [Accepted: 09/03/2024] [Indexed: 09/09/2024]
Abstract
Macrophages are a type of white blood cell that play a significant role in determining the inflammatory response associated with a wide range of medical conditions. They are highly plastic, having the capacity to adopt numerous polarisation states or 'phenotypes' with disparate pro- or anti-inflammatory roles. Many previous studies divide macrophages into two categorisations: M1 macrophages are largely pro-inflammatory in nature, while M2 macrophages are largely restorative. However, there is a growing body of evidence that the M1 and M2 classifications represent the extremes of a much broader spectrum of phenotypes, and that intermediate phenotypes can play important roles in the progression or treatment of many medical conditions. In this article, we present a model of macrophage dynamics that includes a continuous description of phenotype, and hence incorporates intermediate phenotype configurations. We describe macrophage phenotype switching via nonlinear convective flux terms that scale with background levels of generic pro- and anti-inflammatory mediators. Through numerical simulation and bifurcation analysis, we unravel the model's resulting dynamics, paying close attention to the system's multistability and the extent to which key macrophage-mediator interactions provide bifurcations that act as switches between chronic states and restoration of health. We show that interactions that promote M1-like phenotypes generally result in a greater array of stable chronic states, while interactions that promote M2-like phenotypes can promote restoration of health. Additionally, our model admits oscillatory solutions reminiscent of relapsing-remitting conditions, with macrophages being largely polarised toward anti-inflammatory activity during remission, but with intermediate phenotypes playing a role in inflammatory flare-ups. We conclude by reflecting on our observations in the context of the ongoing pursuance of novel therapeutic interventions.
Collapse
Affiliation(s)
- Suliman Almansour
- School of Science & Technology, Nottingham Trent University, Nottingham, NG11 8NS, UK
| | - Joanne L Dunster
- Institute for Cardiovascular and Metabolic Research, University of Reading, Reading, RG6 6AS, UK
| | - Jonathan J Crofts
- School of Science & Technology, Nottingham Trent University, Nottingham, NG11 8NS, UK
| | - Martin R Nelson
- School of Science & Technology, Nottingham Trent University, Nottingham, NG11 8NS, UK.
| |
Collapse
|
29
|
Abbas TF, Ali HZ. Lupeol stimulates iNOS, TNF-α, and IL-10 expression in the U937 cell line infected with old-world Leishmania donovani. Cytokine 2024; 183:156757. [PMID: 39288647 DOI: 10.1016/j.cyto.2024.156757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 09/05/2024] [Accepted: 09/09/2024] [Indexed: 09/19/2024]
Abstract
OBJECTIVE Visceral leishmaniasis is a neglected tropical disease that can be lethal if not treated. The available medicines have severe side effects, such as toxicity and drug resistance. Various investigations are looking into new anti-leishmanial compounds from natural products that have little impact on host cells. Lupeol, a triterpenoid present in the flora of many edible plants, has been shown to have antimicrobial properties. The present study investigated the immunomodulatory effects of lupeol on U937 macrophages infected with Leishmania donovani, focusing on the expression of key cytokines and enzymes involved in the immune response. METHODS U937 macrophages were infected with Leishmania donovani amastigotes and treated with varying concentrations of lupeol throughout three days. The expression levels of inducible nitric oxide synthase (iNOS), tumor necrosis factor-alpha (TNF-α), and interleukin-10 (IL-10) were measured using real-time polymerase chain reaction (RT-PCR). A positive simulation of gene expression was estimated using ΔΔCT to assess relative expression. RESULTS The results demonstrated that lupeol significantly upregulated iNOS and TNF-α expression, especially at higher concentrations, indicating enhanced pro-inflammatory and anti-leishmanial activity. Interestingly, IL-10 expression also increased, suggesting a complex immunomodulatory role of lupeol that involves both pro-inflammatory and anti-inflammatory pathways. Pearson correlation analysis revealed a strong association between iNOS and TNF-α (0.97692), as well as a moderate correlation between iNOS and IL-10 (0.51603). CONCLUSION These findings suggest that lupeol may promote a balanced immune response, enhancing the body's ability to combat L. donovani while potentially mitigating excessive inflammation. Lupeol can potentially serve as a novel therapeutic agent against visceral leishmaniasis.
Collapse
Affiliation(s)
| | - Hayder Z Ali
- Department of Biology, College of Science, University of Baghdad, Al-Jaderiya Campus, Baghdad 10071, Iraq.
| |
Collapse
|
30
|
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
|
31
|
Bhat MF, Srdanović S, Sundberg LR, Einarsdóttir HK, Marjomäki V, Dekker FJ. Impact of HDAC inhibitors on macrophage polarization to enhance innate immunity against infections. Drug Discov Today 2024; 29:104193. [PMID: 39332483 DOI: 10.1016/j.drudis.2024.104193] [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: 05/31/2024] [Revised: 08/26/2024] [Accepted: 09/23/2024] [Indexed: 09/29/2024]
Abstract
Innate immunity plays an important role in host defense against pathogenic infections. It involves macrophage polarization into either the pro-inflammatory M1 or the anti-inflammatory M2 phenotype, influencing immune stimulation or suppression, respectively. Epigenetic changes during immune reactions contribute to long-term innate immunity imprinting on macrophage polarization. It is becoming increasingly evident that epigenetic modulators, such as histone deacetylase (HDAC) inhibitors (HDACi), enable the enhancement of innate immunity by tailoring macrophage polarization in response to immune stressors. In this review, we summarize current literature on the impact of HDACi and other epigenetic modulators on the functioning of macrophages during diseases that have a strong immune component, such as infections. Depending on the disease context and the chosen therapeutic intervention, HDAC1, HDAC2, HDAC3, HDAC6, or HDAC8 are particularly important in influencing macrophage polarization towards either M1 or M2 phenotypes. We anticipate that therapeutic strategies based on HDAC epigenetic mechanisms will provide a unique approach to boost immunity against disease challenges, including resistant infections.
Collapse
Affiliation(s)
- Mohammad Faizan Bhat
- Department of Chemical and Pharmaceutical Biology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, A. Deusinglaan 1, 9713 AV Groningen, the Netherlands
| | - Sonja Srdanović
- Akthelia Pharmaceuticals, Grandagardi 16, 101 Reykjavik, Iceland
| | - Lotta-Riina Sundberg
- Department of Biological and Environmental Sciences and Nanoscience Center, 40014 University of Jyväskylä, Jyväskylä, Finland
| | | | - Varpu Marjomäki
- Department of Biological and Environmental Sciences and Nanoscience Center, 40014 University of Jyväskylä, Jyväskylä, Finland
| | - Frank J Dekker
- Department of Chemical and Pharmaceutical Biology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, A. Deusinglaan 1, 9713 AV Groningen, the Netherlands.
| |
Collapse
|
32
|
Lee SJ, Koh A, Lee SH, Kim KW. Distinct activation of M1 and M2 macrophages in the primary pterygium lymphangiogenesis. Exp Eye Res 2024; 248:110108. [PMID: 39326777 DOI: 10.1016/j.exer.2024.110108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 09/06/2024] [Accepted: 09/23/2024] [Indexed: 09/28/2024]
Abstract
The precise role and innate immunological mechanisms underlying lymphangiogenesis in pterygium remain unclear. This study aimed to investigate the presence of M1 and M2 macrophages and their correlation with pro-lymphangiogenic activation and lymphatic endothelial expression in human pterygium stromal tissues. We analyzed human pterygium and subject-matched normal conjunctival tissues for the expression of these factors and conducted in vitro experiments to assess interactions between macrophages and pterygium fibroblasts. Myeloid and M1 macrophage markers were upregulated in pterygium. M1 macrophages were associated with the upregulation of pro-lymphangiogenic vascular endothelial growth factor C (Vegfc) in pterygium tissues and induced inflammatory signals in pterygium fibroblasts. In contrast, lymphatic vessel endothelial hyaluronan receptor 1 (Lyve1) expression was associated with M2 markers but not with M1 markers. Notably, the clinical severity of pterygium was inversely correlated with the expression of the M2 marker Cd163. These findings suggest that M1 and M2 macrophages play distinct roles in the pathogenesis of pterygium, with M1 macrophages enhancing lymphangiogenic stimulation and inflammatory responses, while M2 macrophages are associated with Lyve1 expression and reduced severity of pterygium. Understanding these mechanisms may advance our current understanding of lymphatic biology in pterygium.
Collapse
Affiliation(s)
- Soo Jin Lee
- Chung-Ang Ocular Surface Restoration via Immune-inflammation Alleviation (CORIA) Laboratory, Seoul, Republic of Korea
| | - Ahra Koh
- Chung-Ang Ocular Surface Restoration via Immune-inflammation Alleviation (CORIA) Laboratory, Seoul, Republic of Korea; Chung-Ang University Graduate School, Seoul, Republic of Korea
| | - Seung Hyeun Lee
- Chung-Ang Ocular Surface Restoration via Immune-inflammation Alleviation (CORIA) Laboratory, Seoul, Republic of Korea; Chung-Ang University Graduate School, Seoul, Republic of Korea; Department of Ophthalmology, Chung-Ang University Gwangmyeong Hospital, Gwangmyeong-si, Gyeonggi-do, Republic of Korea
| | - Kyoung Woo Kim
- Chung-Ang Ocular Surface Restoration via Immune-inflammation Alleviation (CORIA) Laboratory, Seoul, Republic of Korea; Department of Ophthalmology, Chung-Ang University College of Medicine, Chung-Ang University Hospital, Seoul, Republic of Korea.
| |
Collapse
|
33
|
Zhang T, Widdop RE, Ricardo SD. Transition from acute kidney injury to chronic kidney disease: mechanisms, models, and biomarkers. Am J Physiol Renal Physiol 2024; 327:F788-F805. [PMID: 39298548 DOI: 10.1152/ajprenal.00184.2024] [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: 06/26/2024] [Revised: 08/15/2024] [Accepted: 09/01/2024] [Indexed: 09/22/2024] Open
Abstract
Acute kidney injury (AKI) and chronic kidney disease (CKD) are increasingly recognized as interconnected conditions with overlapping pathophysiological mechanisms. This review examines the transition from AKI to CKD, focusing on the molecular mechanisms, animal models, and biomarkers essential for understanding and managing this progression. AKI often progresses to CKD due to maladaptive repair processes, persistent inflammation, and fibrosis, with both conditions sharing common pathways involving cell death, inflammation, and extracellular matrix (ECM) deposition. Current animal models, including ischemia-reperfusion injury (IRI) and nephrotoxic damage, help elucidate these mechanisms but have limitations in replicating the complexity of human disease. Emerging biomarkers such as kidney injury molecule-1 (KIM-1), neutrophil gelatinase-associated lipocalin (NGAL), and soluble tumor necrosis factor receptors (TNFRs) show promise in early detection and monitoring of disease progression. This review highlights the need for improved animal models and biomarker validation to better mimic human disease and enhance clinical translation. Advancing our understanding of the AKI-to-CKD transition through targeted therapies and refined research approaches holds the potential to significantly improve patient outcomes.
Collapse
Affiliation(s)
- Tingfang Zhang
- Department of Pharmacology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Robert E Widdop
- Department of Pharmacology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Sharon D Ricardo
- Department of Pharmacology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
34
|
Chelvanambi S, Decano JL, Winkels H, Giannarelli C, Aikawa M. Decoding Macrophage Heterogeneity to Unravel Vascular Inflammation as a Path to Precision Medicine. Arterioscler Thromb Vasc Biol 2024; 44:2253-2257. [PMID: 39441912 DOI: 10.1161/atvbaha.124.319571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Affiliation(s)
- Sarvesh Chelvanambi
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Department of Medicine (S.C., J.L.D., M.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Julius L Decano
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Department of Medicine (S.C., J.L.D., M.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Holger Winkels
- Faculty of Medicine and University Hospital Cologne, Clinic III for Internal Medicine (H.W.), University of Cologne, Germany
- Center for Molecular Medicine Cologne (H.W.), University of Cologne, Germany
| | - Chiara Giannarelli
- Cardiovascular Research Center, Division of Cardiology, Departments of Medicine and Pathology, New York University Grossman School of Medicine (C.G.)
| | - Masanori Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Department of Medicine (S.C., J.L.D., M.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
- Channing Division of Network Medicine, Department of Medicine (M.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
- Center for Excellence in Vascular Biology (M.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
35
|
Olivieri F, Biscetti L, Pimpini L, Pelliccioni G, Sabbatinelli J, Giunta S. Heart rate variability and autonomic nervous system imbalance: Potential biomarkers and detectable hallmarks of aging and inflammaging. Ageing Res Rev 2024; 101:102521. [PMID: 39341508 DOI: 10.1016/j.arr.2024.102521] [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: 04/10/2024] [Revised: 09/11/2024] [Accepted: 09/23/2024] [Indexed: 10/01/2024]
Abstract
The most cutting-edge issue in the research on aging is the quest for biomarkers that transcend molecular and cellular domains to encompass organismal-level implications. We recently hypothesized the role of Autonomic Nervous System (ANS) imbalance in this context. Studies on ANS functions during aging highlighted an imbalance towards heightened sympathetic nervous system (SNS) activity, instigating a proinflammatory milieu, and attenuated parasympathetic nervous system (PNS) function, which exerts anti-inflammatory effects via the cholinergic anti-inflammatory pathway (CAP) and suppression of the hypothalamic-pituitary-adrenal (HPA) axis. This scenario strongly suggests that ANS imbalance can fuel inflammaging, now recognized as one of the most relevant risk factors for age-related disease development. Recent recommendations have increasingly highlighted the need for actionable strategies to improve the quality of life for older adults by identifying biomarkers that can be easily measured, even in asymptomatic individuals. We advocate for considering ANS imbalance as a biomarker of aging and inflammaging. Measures of ANS imbalance, such as heart rate variability (HRV), are relatively affordable, non-invasive, and cost-effective, making this hallmark easily diagnosable. HRV gains renewed significance within the aging research landscape, offering a tangible link between pathophysiological perturbations and age-related health outcomes.
Collapse
Affiliation(s)
- Fabiola Olivieri
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona, Italy; Advanced Technology Center for Aging Research and Geriatric Mouse Clinic, IRCCS INRCA, Ancona, Italy
| | | | | | | | - Jacopo Sabbatinelli
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona, Italy; Clinic of Laboratory and Precision Medicine, IRCCS INRCA, Ancona, Italy.
| | - Sergio Giunta
- Casa di Cura Prof. Nobili (Gruppo Garofalo GHC), Castiglione dei Pepoli, Bologna, Italy
| |
Collapse
|
36
|
Ho QV, Young MJ. Mineralocorticoid receptors, macrophages and new mechanisms for cardiovascular disease. Mol Cell Endocrinol 2024; 593:112340. [PMID: 39134137 DOI: 10.1016/j.mce.2024.112340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Revised: 08/01/2024] [Accepted: 08/05/2024] [Indexed: 08/16/2024]
Affiliation(s)
- Quoc Viet Ho
- Cardiovascular Endocrinology Laboratory, Baker Heart and Diabetes Institute, Australia
| | - Morag J Young
- Cardiovascular Endocrinology Laboratory, Baker Heart and Diabetes Institute, Australia; Department of Cardiometabolic Health, University of Melbourne, Victoria, Australia.
| |
Collapse
|
37
|
Sun Y, Lian Y, Mei X, Xia J, Feng L, Gao J, Xu H, Zhang X, Yang H, Hao X, Feng Y. Cinobufagin inhibits M2‑like tumor‑associated macrophage polarization to attenuate the invasion and migration of lung cancer cells. Int J Oncol 2024; 65:102. [PMID: 39301639 PMCID: PMC11419410 DOI: 10.3892/ijo.2024.5690] [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: 04/03/2024] [Accepted: 08/14/2024] [Indexed: 09/22/2024] Open
Abstract
Macrophages have crucial roles in immune responses and tumor progression, exhibiting diverse phenotypes based on environmental cues. In the present study, the impact of cinobufagin (CB) on macrophage polarization and the consequences on tumor‑associated behaviors were investigated. Morphological transformations of THP‑1 cells into M0, M1 and M2 macrophages were observed, including distinct changes in the size, shape and adherence properties of these cells. CB treatment inhibited the viability of A549 and LLC cells in a concentration‑dependent manner, with an IC50 of 28.8 and 30.12 ng/ml, respectively. CB at concentrations of <30 ng/ml had no impact on the viability of M0 macrophages and lung epithelial (BEAS‑2B) cells. CB influenced the expression of macrophage surface markers, reducing CD206 positivity in M2 macrophages without affecting CD86 expression in M1 macrophages. CB also altered certain expression profiles at the mRNA level, notably downregulating macrophage receptor with collagenous structure (MARCO) expression in M2 macrophages and upregulating tumor necrosis factor‑α and interleukin‑1β in both M0 and M1 macrophages. Furthermore, ELISA analyses revealed that CB increased the levels of pro‑inflammatory cytokines in M1 macrophages and reduced the levels of anti‑inflammatory factors in M2 macrophages. CB treatment also attenuated the migration and invasion capacities of A549 and LLC cells stimulated by M2 macrophage‑conditioned medium. Additionally, CB modulated peroxisome proliferator‑activated receptor γ (PPARγ) and MARCO expression in M2 macrophages and epithelial‑mesenchymal transition in A549 cells, which was partially reversed by rosiglitazone, a PPARγ agonist. Finally, CB and cisplatin treatments hindered tumor growth in vivo, with distinct impacts on animal body weight and macrophage marker expression in tumor tissues. In conclusion, the results of the present study demonstrated that CB exerted complex regulatory effects on macrophage polarization and tumor progression, suggesting its potential as a modulator of the tumor microenvironment and a therapeutic for cancer treatment.
Collapse
Affiliation(s)
- Ying Sun
- Department of Pathogenic Biology and Immunology, Medical College, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, P.R. China
| | - Yunfeng Lian
- Department of Pathogenic Biology and Immunology, Medical College, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, P.R. China
| | - Xue Mei
- Department of Pathogenic Biology and Immunology, Medical College, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, P.R. China
| | - Jinchan Xia
- Department of Pathogenic Biology and Immunology, Medical College, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, P.R. China
| | - Long Feng
- Department of Pathogenic Biology and Immunology, Medical College, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, P.R. China
| | - Jianfeng Gao
- Department of Pathogenic Biology and Immunology, Medical College, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, P.R. China
| | - Huaming Xu
- Department of Pathogenic Biology and Immunology, Medical College, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, P.R. China
| | - Xiaoyan Zhang
- Department of Epidemic Febrile Disease, Traditional Chinese Medical College, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, P.R. China
| | - Huitong Yang
- Department of Pathogenic Biology and Immunology, Medical College, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, P.R. China
| | - Xu Hao
- Department of Pathogenic Biology and Immunology, Medical College, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, P.R. China
| | - Yilin Feng
- Department of Pathogenic Biology and Immunology, Medical College, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, P.R. China
| |
Collapse
|
38
|
Zhang Y, Rao Y, Lu J, Wang J, Ker DFE, Zhou J, Wang DM. The influence of biophysical niche on tumor-associated macrophages in liver cancer. Hepatol Commun 2024; 8:e0569. [PMID: 39470328 PMCID: PMC11524744 DOI: 10.1097/hc9.0000000000000569] [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: 03/04/2024] [Accepted: 08/30/2024] [Indexed: 10/30/2024] Open
Abstract
HCC, the most common type of primary liver cancer, is a leading cause of cancer-related mortality worldwide. Although the advancement of immunotherapies by immune checkpoint inhibitors (ICIs) that target programmed cell death 1 or programmed cell death 1-ligand 1 has revolutionized the treatment for HCC, the majority is still not beneficial. Accumulating evidence has pointed out that the potent immunosuppressive tumor microenvironment in HCC poses a great challenge to ICI therapeutic efficacy. As a key component in tumor microenvironment, tumor-associated macrophages (TAMs) play vital roles in HCC development, progression, and ICI low responsiveness. Mechanistically, TAM can promote cancer invasion and metastasis, angiogenesis, epithelial-mesenchymal transition, maintenance of stemness, and most importantly, immunosuppression. Targeting TAMs, therefore, represents an opportunity to enhance the ICI therapeutic efficacy in patients with HCC. While previous research has primarily focused on biochemical cues influencing macrophages, emerging evidence highlights the critical role of biophysical signals, such as substrate stiffness, topography, and external forces. In this review, we summarize the influence of biophysical characteristics within the tumor microenvironment that regulate the phenotype and function of TAMs in HCC pathogenesis and progression. We also explore the possible mechanisms and discuss the potential of manipulating biophysical cues in regulating TAM for HCC therapy. By gaining a deeper understanding of how macrophages sense and respond to mechanical forces, we may potentially usher in a path toward a curative approach for combinatory cancer immunotherapies.
Collapse
Affiliation(s)
- Ying Zhang
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China
- Institute of Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China
- Institute of Precision Medicine, Jining Medical University, Jining, China
| | - Ying Rao
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China
- Institute of Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China
| | - Jiahuan Lu
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China
| | - Jiyu Wang
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China
- Institute of Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China
| | - Dai Fei Elmer Ker
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Sha Tin, Hong Kong, SAR, China
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, SAR, China
| | - Jingying Zhou
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China
| | - Dan Michelle Wang
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China
- Institute of Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Sha Tin, Hong Kong, SAR, China
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China
| |
Collapse
|
39
|
Luo M, Zhang W, Yang J, Du X, Wang X, Xu G, Tang H, Wang Z, Zhong X, Feng J, Ma N. CD83 mediates the inhibitory effect of the S1PR1 agonist CYM5442 on LPS-induced M1 polarization of macrophages through the ERK-STAT-1 signaling pathway. Int Immunopharmacol 2024; 143:113526. [PMID: 39486189 DOI: 10.1016/j.intimp.2024.113526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 10/23/2024] [Accepted: 10/25/2024] [Indexed: 11/04/2024]
Abstract
Macrophages possess M1/M2 polarization, which perform an essential role in immunology and inflammation studies. However, few studies have investigated the specific molecules involved in the polarization process beyond its induction and characterization. Here, we determined that the molecule S1PR1 regulates M1 polarization in macrophages and that the surface marker CD83 is involved in this process. The S1PR1 agonist CYM5442 specifically increases CD83 expression in macrophages. Although the agonist CYM5442 and LPS regulate CD83 differently in macrophages, they have a synergistic effect that enhances CD83 expression. Notably, CYM5442 does not act synergistically with IL-4 regarding CD83 expression and does not affect IL-4-induced macrophage M2 polarization. Furthermore, CYM5442 inhibits the expression of LPS-induced inflammatory cytokines and the phosphorylation of ERK1/2 and STAT-1 in macrophages. However, this inhibition was significantly diminished or absent when CD83 is deficient, highlighting the importance of CD83 in mediating S1PR1 signaling in LPS-induced M1 polarization of macrophages. Overall, our findings provide valuable insights into the molecular mechanisms underlying macrophage polarization, particularly the roles of S1PR1 and CD83 in modulating inflammatory responses.
Collapse
Affiliation(s)
- MeiHua Luo
- Inflammation & Allergic Diseases Research Unit, The Affiliated Hospital of Southwest Medical University, Luzhou 646000 Sichuan, China; Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou 646000 Sichuan, China
| | - Wei Zhang
- Inflammation & Allergic Diseases Research Unit, The Affiliated Hospital of Southwest Medical University, Luzhou 646000 Sichuan, China; Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou 646000 Sichuan, China
| | - Juan Yang
- Inflammation & Allergic Diseases Research Unit, The Affiliated Hospital of Southwest Medical University, Luzhou 646000 Sichuan, China; Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou 646000 Sichuan, China
| | - Xi Du
- Laboratory of Anesthesiology, Southwest Medical University, Luzhou 646000 Sichuan, China
| | - Xing Wang
- Inflammation & Allergic Diseases Research Unit, The Affiliated Hospital of Southwest Medical University, Luzhou 646000 Sichuan, China
| | - Guofeng Xu
- Inflammation & Allergic Diseases Research Unit, The Affiliated Hospital of Southwest Medical University, Luzhou 646000 Sichuan, China
| | - Hongmei Tang
- Inflammation & Allergic Diseases Research Unit, The Affiliated Hospital of Southwest Medical University, Luzhou 646000 Sichuan, China
| | - Zhibin Wang
- Inflammation & Allergic Diseases Research Unit, The Affiliated Hospital of Southwest Medical University, Luzhou 646000 Sichuan, China
| | - Xiaolin Zhong
- Department of Gastroenterology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000 Sichuan, China
| | - Jianguo Feng
- Laboratory of Anesthesiology, Southwest Medical University, Luzhou 646000 Sichuan, China.
| | - Ning Ma
- Inflammation & Allergic Diseases Research Unit, The Affiliated Hospital of Southwest Medical University, Luzhou 646000 Sichuan, China.
| |
Collapse
|
40
|
Kricha A, Bouchmaa N, Ben Mkaddem S, Abbaoui A, Ben Mrid R, El Fatimy R. Glioblastoma-associated macrophages: A key target in overcoming glioblastoma therapeutic resistance. Cytokine Growth Factor Rev 2024:S1359-6101(24)00088-1. [PMID: 39510901 DOI: 10.1016/j.cytogfr.2024.10.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 10/24/2024] [Accepted: 10/24/2024] [Indexed: 11/15/2024]
Abstract
Glioblastoma multiforme (GBM) is recognized as the most aggressive and malignant form of brain cancer, characterized by a highly heterogeneous phenotype, poor prognosis, and a median survival time of less than 16 months. Recent studies have highlighted the critical role of glioblastoma-associated macrophages (GAMs) in promoting tumor progression and resistance not only to immunotherapy but also to radiotherapy and chemotherapy. GAMs actively suppress immune responses, promote angiogenesis, facilitate tumor metastasis, and induce therapy resistance, through various mechanisms such as cytokines production, signaling pathways regulation, and angiogenesis. In this context, understanding these regulatory mechanisms is essential for developing efficient therapies. Preclinical studies have investigated diverse approaches to target these cells, both as monotherapies or in combination with other treatments. While these approaches have shown promise in strengthening antitumor immune responses in animal models, their clinical success remains to be fully determined. Herein, we provide a comprehensive overview of GAMs's role in GBM therapeutic resistance and summarizes existing approaches to target GAMs in overcoming tumor resistance.
Collapse
Affiliation(s)
- Aymane Kricha
- Institute of Biological Sciences (IBS), Faculty of Medical Sciences, Mohammed VI Polytechnic University (FMS-UM6P), Benguerir, Morocco.
| | - Najat Bouchmaa
- Institute of Biological Sciences (IBS), Faculty of Medical Sciences, Mohammed VI Polytechnic University (FMS-UM6P), Benguerir, Morocco.
| | - Sanae Ben Mkaddem
- Institute of Biological Sciences (IBS), Faculty of Medical Sciences, Mohammed VI Polytechnic University (FMS-UM6P), Benguerir, Morocco.
| | - Abdellatif Abbaoui
- Institute of Biological Sciences (IBS), Faculty of Medical Sciences, Mohammed VI Polytechnic University (FMS-UM6P), Benguerir, Morocco.
| | - Reda Ben Mrid
- Institute of Biological Sciences (IBS), Faculty of Medical Sciences, Mohammed VI Polytechnic University (FMS-UM6P), Benguerir, Morocco.
| | - Rachid El Fatimy
- Institute of Biological Sciences (IBS), Faculty of Medical Sciences, Mohammed VI Polytechnic University (FMS-UM6P), Benguerir, Morocco.
| |
Collapse
|
41
|
Hu X, Nan Y, Zhang Y, Fan J, Wang H, Bai Y, Zhang Y, Zhang X, Zhu Z, Cao Z, Ye X, Wu T, Xu S, Wu Z, Hu W, Ju D. Simultaneously blocking ANGPTL3 and CD47 prevents the progression of atherosclerosis through regulating lipid metabolism, macrophagic efferocytosis and lipid peroxidation. Pharmacol Res 2024; 210:107486. [PMID: 39488258 DOI: 10.1016/j.phrs.2024.107486] [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: 08/04/2024] [Revised: 10/30/2024] [Accepted: 10/30/2024] [Indexed: 11/04/2024]
Abstract
Atherosclerosis (AS) ultimately cause major adverse cardiovascular events (MACEs). While traditional strategies by lipid-reducing have reduced MACEs, many patients continue to face significant risks. It might attribute to the upregulation of CD47 expression in AS lesions, that mediated anti-efferocytosis of macrophages. Therefore, we propose simultaneously blocking ANGPTL3, a vital regulator of lipid metabolism, and CD47 might be a potential approach for AS therapy. Firstly, we investigate the role of a novel anti-ANGPTL3 nanobody-Fc (FD03) in AS. We found that FD03 treatment significantly decreased circulating lipids, plaque size, and lipid deposition in apoE-/- mice compared to control Ab, but there was a twofold increase in plaque formation in comparison to baseline. However, immunofluorescence indicated the upregulation of CD47 expression in the plaques even after FD03 treatment compared to normal vascular tissue. Next, a bifunctional protein containing signal regulatory protein alpha (SIRPα) and FD03 (SIRPαD1-FD03) was constructed to block CD47 and ANGPTL3 concurrently, which had high purity, robust stability, and high affinity to CD47 and ANGPTL3 with biological activity in vitro. Furthermore, SIRPαD1-FD03 fusion protein exhibited the enhanced therapeutic effect on AS compared with SIRPαD1-Fc or FD03, regressing plaque contents and the necrotic core equal to baseline. Mechanistically, SIRPαD1-FD03 reduced serum lipids, augmented the efferocytosis rate and macrophage M2 polarization, and decreased the reactive oxygen species (ROS) and lipid peroxidation level in atherosclerotic plaques. Collectively, our project suggests an effective approach for AS by simultaneously blocking ANGPTL3 and CD47 to regulate lipid metabolism, macrophage activity and lipid peroxidation.
Collapse
Affiliation(s)
- Xiaozhi Hu
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutic, Fudan University School of Pharmacy, Shanghai, China
| | - Yanyang Nan
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutic, Fudan University School of Pharmacy, Shanghai, China
| | - Yuting Zhang
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutic, Fudan University School of Pharmacy, Shanghai, China
| | - Jiajun Fan
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutic, Fudan University School of Pharmacy, Shanghai, China
| | - Hanqi Wang
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutic, Fudan University School of Pharmacy, Shanghai, China
| | - Yu Bai
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutic, Fudan University School of Pharmacy, Shanghai, China
| | - Yuanzhen Zhang
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutic, Fudan University School of Pharmacy, Shanghai, China
| | - Xuyao Zhang
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutic, Fudan University School of Pharmacy, Shanghai, China
| | - Zeguo Zhu
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutic, Fudan University School of Pharmacy, Shanghai, China
| | - Zhonglian Cao
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutic, Fudan University School of Pharmacy, Shanghai, China
| | - Xiaomiao Ye
- Department of Cardiology, Minhang Hospital, Fudan University, Shanghai, China
| | - Tao Wu
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutic, Fudan University School of Pharmacy, Shanghai, China
| | - Shuwen Xu
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutic, Fudan University School of Pharmacy, Shanghai, China
| | - Zhengyu Wu
- TAU Cambridge Ltd, The Bradfield Centre UNIT 184, Cambridge Science Park, Cambridge CB4 0GA, UK.
| | - Wei Hu
- Department of Cardiology, Minhang Hospital, Fudan University, Shanghai, China.
| | - Dianwen Ju
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutic, Fudan University School of Pharmacy, Shanghai, China; Department of Cardiology, Minhang Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
42
|
Fan W, Liu P, Tan L, Lv H, Zhou H, Tao Z, Xu Y. Tet2 modulates M2 macrophage polarization via mRNA 5-methylcytosine in allergic rhinitis. Int Immunopharmacol 2024; 143:113495. [PMID: 39486186 DOI: 10.1016/j.intimp.2024.113495] [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: 07/30/2024] [Revised: 10/21/2024] [Accepted: 10/22/2024] [Indexed: 11/04/2024]
Abstract
BACKGROUND Allergic rhinitis (AR) represents a hallmark of obvious hypersensitivity with an imbalance of immune responses, including abnormal macrophage activity in local tissues. It has been reported that alternatively activated macrophages (M2) may contribute to allergic pathogenesis. Ten-eleven translocation (Tet) enzymes can oxidize 5-methylcytosine (m5C) in mRNA, implying the epigenetic regulation of post-transcriptional RNA modification. Our previous study suggested that decreased Tet2 impairs the function of regulatory T cells, failing to exert a protective role in AR. However, the mechanism of Tet2 in macrophage polarization has been little discussed. In this paper, we investigate the regulatory role of Tet2 in macrophage polarization under allergic inflammation. METHODS Macrophage immunofluorescence and eosinophil counts were used to confirm the inflammatory and polarized state in the nasal mucosa of AR patients. Additionally, we used Raw264.7 cells to explore the relationships among mRNA methylation, Tet2 expression, and the macrophage polarization process. Furthermore an Ovalbumin (OVA)-mediated AR mouse model was established with wild-type (WT) and Tet2 gene knockout (Tet2-/-) mice to verify the role of Tet2 in AR severity and macrophage polarization. The final stage comprised RNA sequencing, methylated RNA immunoprecipitation with qPCR (MeRIP-qPCR) using bone marrow-derived macrophages (BMDMs) from WT and Tet2-/- mice to explore the effect of Tet2 deficiency on the mRNA methylation level of M2-related genes under OVA treatment. A two-tailed Student's t-test was used to compare two groups, and Spearman correlation analysis was applied for relationship analysis. RESULTS M2-macrophages were confirmed as the dominant subtype associated with eosinophil levels in AR nasal tissues. In vitro analyses demonstrated that mRNA methylation and Tet2 are linked to M2 macrophages. Additionally, we found that Tet2 influences local allergic severity and macrophage polarization. Specifically, Tet2 deficiency decreased the mRNA m5C demethylation levels of Klf4 and Rock1, contributing to M2 polarization in an allergic state. CONCLUSIONS The findings of this study demonstrate that Tet2 may play a protective role in AR by negatively regulating M2-related factors through mRNA m5C demethylation. These findings provide new insights into AR therapy, suggesting that intervening in macrophage polarization at the post-transcriptional level could be a novel therapeutic strategy.
Collapse
Affiliation(s)
- Wenjun Fan
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Peiqiang Liu
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lu Tan
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Hao Lv
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Huiqin Zhou
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zezhang Tao
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yu Xu
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
43
|
Shan X, Ji Z, Wang B, Zhang Y, Dong H, Jing W, Zhou Y, Hu P, Cui Y, Li Z, Yu S, Zhou J, Wang T, Shen L, Liu Y, Yu Q. Riboflavin kinase binds and activates inducible nitric oxide synthase to reprogram macrophage polarization. Redox Biol 2024; 78:103413. [PMID: 39536592 DOI: 10.1016/j.redox.2024.103413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 10/24/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
Riboflavin kinase (RFK) is essential in riboflavin metabolism, converting riboflavin to flavin mononucleotide (FMN), which is further processed to flavin adenine dinucleotide (FAD). While RFK enhances macrophage phagocytosis of Listeria monocytogenes, its role in macrophage polarization is not well understood. Our study reveals that RFK deficiency impairs M(IFN-γ) and promotes M(IL-4) polarization, both in vitro and in vivo. Mechanistically, RFK interacts with inducible nitric oxide (NO) synthase (iNOS), which requires FMN and FAD as cofactors for activation, leading to increased NO production that alters energy metabolism by inhibiting the tricarboxylic acid cycle and mitochondrial electron transport chain. Exogenous FAD reverses the metabolic and polarization changes caused by RFK deficiency. Furthermore, bone marrow adoptive transfer from high-riboflavin-fed mice into wild-type tumor-bearing mice reprograms tumor-associated macrophage polarization and inhibits tumor growth. These results suggest that targeting RFK-iNOS or modulating riboflavin metabolism could be potential therapies for macrophage-related immune diseases.
Collapse
Affiliation(s)
- Xiao Shan
- Department of Health Management Centre & Institute of Health Management, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610000, China; Tianjin Institute of Immunology, Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory of Inflammation Biology, State Key Laboratory of Experimental Hematology, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China.
| | - Zemin Ji
- Tianjin Institute of Immunology, Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory of Inflammation Biology, State Key Laboratory of Experimental Hematology, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Baochen Wang
- Tianjin Institute of Immunology, Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory of Inflammation Biology, State Key Laboratory of Experimental Hematology, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Yanan Zhang
- Tianjin Institute of Immunology, Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory of Inflammation Biology, State Key Laboratory of Experimental Hematology, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Hongyuan Dong
- Tianjin Institute of Immunology, Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory of Inflammation Biology, State Key Laboratory of Experimental Hematology, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Weijia Jing
- Tianjin Institute of Immunology, Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory of Inflammation Biology, State Key Laboratory of Experimental Hematology, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Yanzhao Zhou
- Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou 450008, China; University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Penghui Hu
- Department of Critical Care Medicine, Department of Anesthesiology, Tianjin Institute of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Yan Cui
- Tianjin Institute of Immunology, Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory of Inflammation Biology, State Key Laboratory of Experimental Hematology, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Zihan Li
- Tianjin Institute of Immunology, Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory of Inflammation Biology, State Key Laboratory of Experimental Hematology, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Sujun Yu
- Tianjin Institute of Immunology, Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory of Inflammation Biology, State Key Laboratory of Experimental Hematology, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Jinxue Zhou
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou 450008, China
| | - Ting Wang
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, School of Basic Medical Sciences, Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, Tianjin Medical University, Tianjin 300070, China
| | - Long Shen
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China; Tianjin Institutes of Health Science, Tianjin 301600, China.
| | - Yuping Liu
- Department of Health Management Centre & Institute of Health Management, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610000, China.
| | - Qiujing Yu
- Department of Health Management Centre & Institute of Health Management, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610000, China.
| |
Collapse
|
44
|
Xu W, Hou H, Yang W, Tang W, Sun L. Immunologic role of macrophages in sepsis-induced acute liver injury. Int Immunopharmacol 2024; 143:113492. [PMID: 39471696 DOI: 10.1016/j.intimp.2024.113492] [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: 08/15/2024] [Revised: 10/11/2024] [Accepted: 10/21/2024] [Indexed: 11/01/2024]
Abstract
Sepsis-induced acute liver injury (SALI), a manifestation of sepsis multi-organ dysfunction syndrome, is associated with poor prognosis and high mortality. The diversity and plasticity of liver macrophage subpopulations explain their different functional responses in different liver diseases. Kupffer macrophages, liver capsular macrophages, and monocyte-derived macrophages are involved in pathogen recognition and clearance and in the regulation of inflammatory responses, exacerbating the progression of SALI through different pathways of pyroptosis, ferroptosis, and autophagy. Concurrently, they play an important role in maintaining hepatic homeostasis and in the injury and repair processes of SALI. Other macrophages are recruited to diseased tissues under pathological conditions and are polarized into various phenotypes (mainly M1 and M2 types) under the influence of signaling molecules, transcription factors, and metabolic reprogramming, thereby exerting different roles and functions. This review provides an overview of the immune role of macrophages in SALI and discusses the multiple roles of macrophages in liver injury and repair to provide a reference for future studies.
Collapse
Affiliation(s)
- Wanling Xu
- Department of Emergency, Jilin University First Hospital, 71 Xinmin Street, Changchun 130021, Jilin, China
| | - Hailong Hou
- Emergency Department, Meihekou Central Hospital, 2668 Aimin Street, Tonghua 135000, Jilin, China
| | - Weiying Yang
- Department of Emergency, Jilin University First Hospital, 71 Xinmin Street, Changchun 130021, Jilin, China
| | - Wenjing Tang
- Department of Emergency, Jilin University First Hospital, 71 Xinmin Street, Changchun 130021, Jilin, China
| | - Lichao Sun
- Department of Emergency, Jilin University First Hospital, 71 Xinmin Street, Changchun 130021, Jilin, China.
| |
Collapse
|
45
|
Zhang D, Shi C, Wang Y, Guo J, Gong Z. Metabolic Dysregulation and Metabolite Imbalances in Acute-on-chronic Liver Failure: Impact on Immune Status. J Clin Transl Hepatol 2024; 12:865-877. [PMID: 39440217 PMCID: PMC11491507 DOI: 10.14218/jcth.2024.00203] [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: 06/18/2024] [Revised: 08/26/2024] [Accepted: 09/04/2024] [Indexed: 10/25/2024] Open
Abstract
Liver failure encompasses a range of severe clinical syndromes resulting from the deterioration of liver function, triggered by factors both within and outside the liver. While the definition of acute-on-chronic liver failure (ACLF) may vary by region, it is universally recognized for its association with multiorgan failure, a robust inflammatory response, and high short-term mortality rates. Recent advances in metabolomics have provided insights into energy metabolism and metabolite alterations specific to ACLF. Additionally, immunometabolism is increasingly acknowledged as a pivotal mechanism in regulating immune cell functions. Therefore, understanding the energy metabolism pathways involved in ACLF and investigating how metabolite imbalances affect immune cell functionality are crucial for developing effective treatment strategies for ACLF. This review methodically examined the immune and metabolic states of ACLF patients and elucidated how alterations in metabolites impact immune functions, offering novel perspectives for immune regulation and therapeutic management of liver failure.
Collapse
Affiliation(s)
- Danmei Zhang
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Chunxia Shi
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Yukun Wang
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Jin Guo
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Zuojiong Gong
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
46
|
Byun KA, Seo SB, Oh S, Jang JW, Son KH, Byun K. Poly-D,L-Lactic Acid Filler Attenuates Ultraviolet B-Induced Skin Pigmentation by Reducing Destruction of the Basement Membrane. Int J Mol Sci 2024; 25:11568. [PMID: 39519119 PMCID: PMC11546917 DOI: 10.3390/ijms252111568] [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: 09/25/2024] [Revised: 10/19/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
Poly-D,L-lactic acid (PDLLA) filler, which increases volume and collagen synthesis, is used for skin rejuvenation. PDLLA filler also increases M2 macrophages and IL-10. Ultraviolet (UV) radiation induces dermal hyperpigmentation by disrupting the basement membrane (BM), allowing melanin to move into the dermis. Therefore, using UV-irradiated macrophages and animal skin, we determined whether PDLLA filler decreased M1 macrophages and skin inflammation, thereby reducing BM destruction and dermal hyperpigmentation. UV radiation increased the M1 macrophage marker CD86 and TNF-α expression, which was inhibited by the treatment of macrophages with PDLLA. In fibroblasts treated with conditioned medium from UV-irradiated macrophages, NF-κB activity, NLRP3 inflammasome components (NLRP3, ASC, and pro-caspase-1), IL-18, MMP2, and MMP9 increased, but all decreased after PDLLA treatment. Similar to the in vitro study, UV-irradiated mouse skin showed increased CD86, NLRP3, ASC, pro-caspase-1, MMP2, and MMP9, which decreased after PDLLA injection. Disruption of the lamina densa of the BM and dermal pigmentation increased after UV irradiation and decreased after PDLLA injection. In conclusion, PDLLA reduced dermal pigmentation by decreasing BM destruction in UV-irradiated skin. PDLLA has the potential to reduce dermal pigmentation by regenerating the BM.
Collapse
Affiliation(s)
- Kyung-A Byun
- Department of Anatomy & Cell Biology, College of Medicine, Gachon University, Incheon 21936, Republic of Korea
- LIBON Inc., Incheon 22006, Republic of Korea
- Functional Cellular Networks Laboratory, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Republic of Korea
| | - Suk Bae Seo
- SeoAh Song Dermatologic Clinic, Seoul 05557, Republic of Korea
| | - Seyeon Oh
- Functional Cellular Networks Laboratory, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Republic of Korea
| | - Jong-Won Jang
- Functional Cellular Networks Laboratory, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Republic of Korea
- Department of Health Sciences and Technology, Gachon Advanced Institute for Health & Sciences and Technology (GAIHST), Gachon University, Incheon 21999, Republic of Korea
| | - Kuk Hui Son
- Department of Thoracic and Cardiovascular Surgery, Gachon University Gil Medical Center, Gachon University, Incheon 21565, Republic of Korea
| | - Kyunghee Byun
- Department of Anatomy & Cell Biology, College of Medicine, Gachon University, Incheon 21936, Republic of Korea
- Functional Cellular Networks Laboratory, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Republic of Korea
- Department of Health Sciences and Technology, Gachon Advanced Institute for Health & Sciences and Technology (GAIHST), Gachon University, Incheon 21999, Republic of Korea
| |
Collapse
|
47
|
Zhang Y, Zou M, Wu H, Zhu J, Jin T. The cGAS-STING pathway drives neuroinflammation and neurodegeneration via cellular and molecular mechanisms in neurodegenerative diseases. Neurobiol Dis 2024; 202:106710. [PMID: 39490400 DOI: 10.1016/j.nbd.2024.106710] [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: 07/03/2024] [Revised: 09/27/2024] [Accepted: 10/18/2024] [Indexed: 11/05/2024] Open
Abstract
Neurodegenerative diseases (NDs) are a type of common chronic progressive disorders characterized by progressive damage to specific cell populations in the nervous system, ultimately leading to disability or death. Effective treatments for these diseases are still lacking, due to a limited understanding of their pathogeneses, which involve multiple cellular and molecular pathways. The triggering of an immune response is a common feature in neurodegenerative disorders. A critical challenge is the intricate interplay between neuroinflammation, neurodegeneration, and immune responses, which are not yet fully characterized. In recent years, the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon gene (STING) pathway, a crucial immune response for intracellular DNA sensing, has gradually gained attention. However, the specific roles of this pathway within cellular types such as immune cells, glial and neuronal cells, and its contribution to ND pathogenesis, remain not fully elucidated. In this review, we systematically explore how the cGAS-STING signaling links various cell types with related cellular effector pathways under the context of NDs for multifaceted therapeutic directions. We emphasize the discovery of condition-dependent cellular heterogeneity in the cGAS-STING pathway, which is integral for understanding the diverse cellular responses and potential therapeutic targets. Additionally, we review the pathogenic role of cGAS-STING activation in Parkinson's disease, ataxia-telangiectasia, and amyotrophic lateral sclerosis. We focus on the complex bidirectional roles of the cGAS-STING pathway in Alzheimer's disease, Huntington's disease, and multiple sclerosis, revealing their double-edged nature in disease progression. The objective of this review is to elucidate the pivotal role of the cGAS-STING pathway in ND pathogenesis and catalyze new insights for facilitating the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Yuxin Zhang
- Department of Neurology, Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Meijuan Zou
- Department of Neurology, Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Hao Wu
- Department of Neurology, Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Jie Zhu
- Department of Neurology, Neuroscience Center, The First Hospital of Jilin University, Changchun, China; Department of Neurobiology, Care Sciences & Society, Karolinska Institute, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Tao Jin
- Department of Neurology, Neuroscience Center, The First Hospital of Jilin University, Changchun, China.
| |
Collapse
|
48
|
Alotiby A. Immunology of Stress: A Review Article. J Clin Med 2024; 13:6394. [PMID: 39518533 PMCID: PMC11546738 DOI: 10.3390/jcm13216394] [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: 09/25/2024] [Revised: 10/19/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024] Open
Abstract
Stress significantly impacts the immune system, affecting susceptibility to illness and overall health. This review examines the intricate relationship between stress and the immune system, offering insights having practical implications for health and disease prevention. Stress can significantly trigger molecular and immune modulation, affecting the distribution and trafficking of immune cells in various organs and altering their composition in the blood. The review delves into two key pathways connecting stress and immunity: the hypothalamic-pituitary-adrenal (HPA) axis and the sympathetic nervous system. Stress activates the neuroendocrine system and triggers microglia in the brain, releasing stress hormones and neurotransmitters that modulate the function and movement of immune cells. Acute stress can temporarily strengthen immunity and promote protection during infection; in contrast, chronic stress dysregulates or inhibits immune functions. Chronic stress causes an increase in cortisol levels through the HPA axis, ultimately suppressing the immune response. Recognizing stress triggers and implementing effective stress management techniques can significantly impact individuals' well-being. This review indicates that immune cells express genes differentially in response to stress, suggesting individual variabilities in the immune response against stress. This underscores the need for a personalized approach to stress management. This review also highlights the potential link between chronic stress and autoimmune disorders and warrants further investigation.
Collapse
Affiliation(s)
- Amna Alotiby
- Department of Hematology and Immunology, Faculty of Medicine, Umm Al-Qura University, Makkah 24381, Saudi Arabia
| |
Collapse
|
49
|
Xiong J, Zhou X, Su L, Jiang L, Ming Z, Pang C, Fuller C, Xu K, Chi H, Zheng X. The two-sided battlefield of tumour-associated macrophages in glioblastoma: unravelling their therapeutic potential. Discov Oncol 2024; 15:590. [PMID: 39453528 PMCID: PMC11511804 DOI: 10.1007/s12672-024-01464-5] [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: 08/09/2024] [Accepted: 10/15/2024] [Indexed: 10/26/2024] Open
Abstract
Gliomas are the most common primary malignant tumours of the central nervous system (CNS), which are highly aggressive, with increasing morbidity and mortality rates year after year, posing a serious threat to the quality and expected survival time of patients. The treatment of gliomas is a major challenge in the field of neuro-oncology, especially high-grade gliomas such as glioblastomas (GBMs). Despite considerable progress in recent years in the study of the molecular and cellular mechanisms of GBMs, their prognosis remains bleak. Tumour-associated macrophages (TAMs) account for up to 50% of GBMs, and they are a highly heterogeneous cell population whose role cannot be ignored. Here, we focus on reviewing the contribution of classically activated M1-phenotype TAMs and alternatively activated M2-phenotype TAMs to GBMs, and exploring the research progress in reprogramming M1 TAMs into M2 TAMs.
Collapse
Affiliation(s)
- Jingwen Xiong
- Department of Sports Rehabilitation, Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Xuancheng Zhou
- Clinical Medical College, Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Lanqian Su
- Clinical Medical College, Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Lai Jiang
- Clinical Medical College, Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Ziwei Ming
- Department of Sports Rehabilitation, Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Can Pang
- School of Public Health, Lanzhou University, Lanzhou, 730000, China
| | - Claire Fuller
- Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, 21224, USA
| | - Ke Xu
- Department of Oncology, Chongqing General Hospital, Chongqing University, Chongqing, 401147, China.
| | - Hao Chi
- Clinical Medical College, Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China.
| | - Xiaomei Zheng
- Department of Neurology, Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China.
| |
Collapse
|
50
|
Seo J, Ha G, Lee G, Nasiri R, Lee J. Modeling tumor-immune interactions using hybrid spheroids and microfluidic platforms for studying tumor-associated macrophage polarization in melanoma. Acta Biomater 2024:S1742-7061(24)00629-9. [PMID: 39461691 DOI: 10.1016/j.actbio.2024.10.036] [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: 07/16/2024] [Revised: 10/19/2024] [Accepted: 10/23/2024] [Indexed: 10/29/2024]
Abstract
Tumor-associated macrophages (TAMs), as key components of tumor microenvironment (TME), exhibit phenotypic plasticity in response to environmental cues, causing polarization into either pro-inflammatory M1 phenotypes or immunosuppressive M2 phenotypes. Although TAM has been widely studied for its crucial involvement in the initiation, progression, metastasis, and immune regulation of cancer cells, there have been limited attempts to understand how the metastatic potentials of cancer cells influence TAM polarization within TME. Here, we developed a miniaturized TME model using a 3D hybrid system composed of murine melanoma cells and macrophages, aiming to investigate interactions between cancer cells exhibiting various metastatic potentials and macrophages within TME. The increase in spheroid size within this model was associated with a reduction in cancer cell viability. Examining macrophage surface marker expression and cytokine secretion indicated the development of diverse TMEs influenced by both spheroid size and the metastatic potential of cancer cells. Furthermore, a high-throughput microfluidic platform equipped with trapping systems and hybrid spheroids was employed to simulate the tumor-immune system of complex TMEs and for comparative analysis with traditional 3D culture models. This study provides insight into TAM polarization in melanoma with different heterogeneities by modeling cancer-immune systems, which can be potentially employed for immune-oncology research, drug screening, and personalized therapy. STATEMENT OF SIGNIFICANCE: This study presents the development of a 3D hybrid spheroid system designed to model tumor-immune interactions, providing a detailed analysis of how melanoma cell metastatic potential influences tumor-associated macrophage (TAM) polarization. By utilizing a microfluidic platform, we are able to replicate and investigate the complex tumor-immune system of the tumor microenvironments (TMEs) under continuous flow conditions. Our model holds significant potential for high-throughput drug screening and personalized medicine applications, offering a versatile tool for advancing cancer research and treatment strategies.
Collapse
Affiliation(s)
- Junki Seo
- Division of Interdisciplinary Bioscience & Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang 790-784, Republic of Korea
| | - Giheon Ha
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), Pohang 790-784, Republic of Korea
| | - Geonho Lee
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), Pohang 790-784, Republic of Korea
| | - Rohollah Nasiri
- Department of Radiation Oncology, Stanford University, Stanford, CA 94305, United States
| | - Junmin Lee
- Division of Interdisciplinary Bioscience & Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang 790-784, Republic of Korea; Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), Pohang 790-784, Republic of Korea; Institute for Convergence Research and Education in Advanced Technology, Yonsei University, Incheon 21983, Republic of Korea.
| |
Collapse
|