1
|
Chen S, Jiang Y, Chai X, Chen Z, Tian H, Liu M, Zhu T, ShangGuan W, Wu X. Uterine-derived exosomes induce the M2 polarization of macrophages via miR-210-3p/ATP5D to promote endometriosis progression. Life Sci 2025; 363:123383. [PMID: 39798647 DOI: 10.1016/j.lfs.2025.123383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 01/06/2025] [Accepted: 01/07/2025] [Indexed: 01/15/2025]
Abstract
AIMS Endometriosis development is associated with peritoneal immune microenvironment abnormality; however, the specific mechanism is uncertain. We aimed to investigate the effects and underlying mechanisms of uterine cavity-derived exosomes on macrophage polarization and endometriosis progression. MATERIALS AND METHODS Uterine cavity-derived exosomes, miR-210-3p inhibitor or siATP5D were used to treat macrophages. Then evaluated the polarization of macrophages. By co-culturing of treated macrophages with endometrial stromal cells in vitro and an endometriosis C57BL6 mouse model to assess the role of uterine-derived exosomes and macrophages in the development of endometriosis. KEY FINDINGS Uterine cavity-derived exosomes could increase miR-210-3p expression and induce M2 macrophage polarization. Mechanistically, miR-210-3p can restrict ATP5D expression in macrophages, which leads to M2 polarization. In vivo experiments confirmed that macrophages lentivirally transduced with miR-210-3p can significantly decrease the growth and implantation of mouse endometriosis. SIGNIFICANCE In summary, our findings suggest that exosomes derived from the uterine cavity may drive macrophages towards M2 and promote endometriosis progression via miR-210-3p/ATP5D.
Collapse
Affiliation(s)
- Shengnan Chen
- Department of Obstetrics and Gynecology, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Ying Jiang
- Department of Obstetrics and Gynecology, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Xiaoshan Chai
- Department of Obstetrics and Gynecology, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Zhaoying Chen
- Department of Obstetrics and Gynecology, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Hao Tian
- Department of Obstetrics and Gynecology, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Min Liu
- Department of Obstetrics and Gynecology, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Tianyu Zhu
- Department of Obstetrics and Gynecology, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Wanwan ShangGuan
- Department of Obstetrics and Gynecology, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Xianqing Wu
- Department of Obstetrics and Gynecology, The Second Xiangya Hospital of Central South University, Changsha 410011, China.
| |
Collapse
|
2
|
Rodríguez JP, Casas J, Balboa MA, Balsinde J. Bioactive lipid signaling and lipidomics in macrophage polarization: Impact on inflammation and immune regulation. Front Immunol 2025; 16:1550500. [PMID: 40028333 PMCID: PMC11867965 DOI: 10.3389/fimmu.2025.1550500] [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: 12/23/2024] [Accepted: 01/28/2025] [Indexed: 03/05/2025] Open
Abstract
Macrophages, crucial innate immune cells, defend against pathogens and resolve inflammation, maintaining tissue balance. They perform phagocytosis, present antigens to T cells, and bond innate and adaptive immunity through various activation states. Classical activation is associated with Th1 responses and interferon γ production, while alternative activation, induced by interleukin 4, is characterized by increased endocytosis, reduced secretion of pro-inflammatory cytokines, and roles in immunoregulation and tissue remodeling. Although these represent opposite extremes observed in vitro, the remarkable plasticity of macrophages allows for a wide spectrum of activation phenotypes that are complex to characterize experimentally. While the application of omics techniques has resulted in significant advances in the characterization of macrophage polarization, lipidomic studies have received lesser attention. Beyond their role as structural components and energy sources, lipids function as signaling molecules that regulate macrophage activation and polarization, thereby shaping immune responses. This work reviews the interaction between lipid signaling and macrophage polarization, exploring how lipid metabolism influences macrophage phenotype and function. These insights offer potential therapeutic strategies for immune-mediated diseases and inflammation-related disorders, including inflammaging.
Collapse
Affiliation(s)
- Juan P. Rodríguez
- Laboratorio de Investigaciones Bioquímicas de la Facultad de Medicina (LIBIM), Instituto de Química Básica y Aplicada del Nordeste Argentino (IQUIBA-NEA), Universidad Nacional del Nordeste, Consejo Nacional de Investigaciones Científicas y Técnicas (UNNE-CONICET), Corrientes, Argentina
| | - Javier Casas
- Instituto de Biología y Genética Molecular, Consejo Superior de Investigaciones Científicas Uva, Valladolid, Spain
- Departamento de Bioquímica y Biología Molecular y Fisiología, Universidad de Valladolid, Valladolid, Spain
| | - María A. Balboa
- Instituto de Biología y Genética Molecular, Consejo Superior de Investigaciones Científicas Uva, Valladolid, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Jesús Balsinde
- Instituto de Biología y Genética Molecular, Consejo Superior de Investigaciones Científicas Uva, Valladolid, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
3
|
Zhai Z, Yang C, Yin W, Liu Y, Li S, Ye Z, Xie M, Song X. Engineered Strategies to Interfere with Macrophage Fate in Myocardial Infarction. ACS Biomater Sci Eng 2025; 11:784-805. [PMID: 39884780 DOI: 10.1021/acsbiomaterials.4c02061] [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: 02/01/2025]
Abstract
Myocardial infarction (MI), a severe cardiovascular condition, is typically triggered by coronary artery disease, resulting in ischemic damage and the subsequent necrosis of the myocardium. Macrophages, known for their remarkable plasticity, are capable of exhibiting a range of phenotypes and functions as they react to diverse stimuli within their local microenvironment. In recent years, there has been an increasing number of studies on the regulation of macrophage behavior based on tissue engineering strategies, and its regulatory mechanisms deserve further investigation. This review first summarizes the effects of key regulatory factors of engineered biomaterials (including bioactive molecules, conductivity, and some microenvironmental factors) on macrophage behavior, then explores specific methods for inducing macrophage behavior through tissue engineering materials to promote myocardial repair, and summarizes the role of macrophage-host cell crosstalk in regulating inflammation, vascularization, and tissue remodeling. Finally, we propose some future challenges in regulating macrophage-material interactions and tailoring personalized biomaterials to guide macrophage phenotypes.
Collapse
Affiliation(s)
- Zitong Zhai
- Central Laboratory, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong 510910, China
| | - Chang Yang
- Central Laboratory, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong 510910, China
| | - Wenming Yin
- Department of Neurology, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong 510910, China
| | - Yali Liu
- Department of Neurology, Foshan Hospital of Traditional Chinese Medicine, Foshan, Guangdong 528000, China
| | - Shimin Li
- Central Laboratory, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong 510910, China
| | - Ziyi Ye
- Central Laboratory, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong 510910, China
| | - Mingxiang Xie
- Central Laboratory, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong 510910, China
| | - Xiaoping Song
- Central Laboratory, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong 510910, China
- Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou, Guangdong 510515, China
| |
Collapse
|
4
|
Liu X, Wang T, Xiang R, Sun H, Zhao M, Ye X, Zhou Y, Wang G, Zhou Y. Anti-inflammatory effects of 1,7-dihydroxy-3,4-dimethoxyxanthone through inhibition of M1-phenotype macrophages via arginine/mitochondrial axis. Immunol Res 2024; 72:1404-1416. [PMID: 39349673 DOI: 10.1007/s12026-024-09538-w] [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: 06/04/2024] [Accepted: 09/02/2024] [Indexed: 02/06/2025]
Abstract
It is known that 1,7-dihydroxy-3,4-dimethoxyxanthone (XAN), derived from Securidaca inappendiculata Hassk., exhibits anti-inflammatory and analgesic activities and inhibits M1 polarization of macrophages. However, its ability to alleviate inflammation induced by pro-inflammatory cytokines in THP-1 cells and its anti-inflammatory mechanisms remain unclear. THP-1 cells were treated with phorbol 12-myristate-13-acetate to differentiate and divided into three groups. They were stimulated with lipopolysaccharide (LPS) and interferon-γ (IFN-γ). The toxicity of XAN was assessed using Cell Counting Kit-8, and the expression of various genes and proteins was analyzed using real-time quantitative polymerase chain reaction, flow cytometry, and western blotting. Transmission electron microscopy was used to observe changes in mitochondrial structure. XAN at concentrations ≤ 10 µg/mL did not affect THP-1 cell viability and reduced the mRNA expression of pro-inflammatory factors, including interleukin (IL)-1β, inducible nitric oxide synthase (iNOS), NOD-like receptor thermal protein domain protein 3 (NLRP3), and tumor necrosis factor-α (TNF-α). XAN also increased the levels of anti-inflammatory factors, including chemokine ligand 22, mannose receptor (CD206), IL-10, peroxisome proliferator-activated receptor-γ, and transglutaminase 2. Additionally, XAN downregulated the expression of inflammation-related proteins iNOS, NLRP3, and IL-1β; significantly increased the expression of arginase 1, ornithine decarboxylase, and arginine metabolism-related proteins and genes; inhibited mitochondrial damage; and reduced reactive oxygen species (ROS) generation. XAN enhanced the arginine metabolism pathway, prevented mitochondrial damage, reduced ROS levels, and provided an effective defensive response against LPS/IFN-γ-induced inflammation.
Collapse
Affiliation(s)
- Xin Liu
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, Wannan Medical College, Wuhu, 241002, China
- Anhui Provincial Engineering Research Center for Polysaccharide Drugs, Wannan Medical College, Wuhu, 241002, China
| | - Ting Wang
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, Wannan Medical College, Wuhu, 241002, China
- Anhui Provincial Engineering Research Center for Polysaccharide Drugs, Wannan Medical College, Wuhu, 241002, China
| | - Ruoxuan Xiang
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, Wannan Medical College, Wuhu, 241002, China
- Anhui Provincial Engineering Research Center for Polysaccharide Drugs, Wannan Medical College, Wuhu, 241002, China
| | - Huazhan Sun
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, Wannan Medical College, Wuhu, 241002, China
- Anhui Provincial Engineering Research Center for Polysaccharide Drugs, Wannan Medical College, Wuhu, 241002, China
| | - Mengyan Zhao
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, Wannan Medical College, Wuhu, 241002, China
- Anhui Provincial Engineering Research Center for Polysaccharide Drugs, Wannan Medical College, Wuhu, 241002, China
| | - Xiaojuan Ye
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, Wannan Medical College, Wuhu, 241002, China
- Anhui Provincial Engineering Research Center for Polysaccharide Drugs, Wannan Medical College, Wuhu, 241002, China
| | - Yuyun Zhou
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, Wannan Medical College, Wuhu, 241002, China
- Anhui Provincial Engineering Research Center for Polysaccharide Drugs, Wannan Medical College, Wuhu, 241002, China
| | - Guodong Wang
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, Wannan Medical College, Wuhu, 241002, China.
- Anhui Provincial Engineering Research Center for Polysaccharide Drugs, Wannan Medical College, Wuhu, 241002, China.
- Anhui Provincial Engineering Laboratory for Screening and Re-Evaluation of Active Compounds of Herbal Medicines in Southern Anhui, Wannan Medical College, Wuhu, 241002, China.
- Anhui Provincial Key Laboratory of Active Biological Macromolecules, Wannan Medical College, Wuhu, 241002, China.
| | - Yuyan Zhou
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, Wannan Medical College, Wuhu, 241002, China.
- Anhui Provincial Engineering Research Center for Polysaccharide Drugs, Wannan Medical College, Wuhu, 241002, China.
- Anhui Provincial Engineering Laboratory for Screening and Re-Evaluation of Active Compounds of Herbal Medicines in Southern Anhui, Wannan Medical College, Wuhu, 241002, China.
- Anhui Provincial Key Laboratory of Active Biological Macromolecules, Wannan Medical College, Wuhu, 241002, China.
| |
Collapse
|
5
|
Kong Y, Zhang Q, Wang S, Li R, Fu C, Wei Q. Mitochondrial metabolism regulated macrophage phenotype in myocardial infarction. Biomed Pharmacother 2024; 180:117494. [PMID: 39321509 DOI: 10.1016/j.biopha.2024.117494] [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: 06/02/2024] [Revised: 09/09/2024] [Accepted: 09/20/2024] [Indexed: 09/27/2024] Open
Abstract
Cardiovascular disease (CVD) remains the leading cause of death worldwide, with myocardial infarction (MI) being the primary contributor to mortality and disability associated with CVD. Reperfusion therapies are widely recognized as effective strategies for treating MI. However, while intended to restore blood flow, the reperfusion processes paradoxically initiate a series of pathophysiological events that worsen myocardial injury, resulting in ischemia-reperfusion (I/R) injury. Therefore, there is a pressing need for new treatment strategies to reduce the size of MI and enhance cardiac function post-infarction. Macrophages are crucial for maintaining homeostasis and mitigating undesirable remodeling following MI. Extensive research has established a strong link between cellular metabolism and macrophage function. In the context of MI, macrophages undergo adaptive metabolic reprogramming to mount an immune response. Moreover, mitochondrial metabolism in macrophages is evident, leading to significant changes in their metabolism. Therefore, we need to delve deeper into summarizing and understanding the relationship and role between mitochondrial metabolism and macrophage phenotype, and summarize existing treatment methods. In this review, we explore the role of mitochondria in shaping the macrophage phenotype and function. Additionally, we summarize current therapeutic strategies aimed at modulating mitochondrial metabolism of macrophages, which may offer new insights treating of MI.
Collapse
Affiliation(s)
- Youli Kong
- Department of Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China; Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, Sichuan, PR China
| | - Qing Zhang
- Department of Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China; Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, Sichuan, PR China
| | - Shiqi Wang
- Department of Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China; Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, Sichuan, PR China
| | - Ran Li
- Department of Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China; Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, Sichuan, PR China
| | - Chenying Fu
- State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Aging and Geriatric Mechanism Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China
| | - Quan Wei
- Department of Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China; Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, Sichuan, PR China.
| |
Collapse
|
6
|
Zhao W, Ye T, Zhou J, Zhang X, Wang K, Zhang H, Cui J, Zhang S, Wang L. Hydrogen bonding-mediated interaction underlies the enhanced membrane toxicity of chemically transformed polystyrene microplastics by cadmium. JOURNAL OF HAZARDOUS MATERIALS 2024; 478:135562. [PMID: 39178779 DOI: 10.1016/j.jhazmat.2024.135562] [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: 02/27/2024] [Revised: 08/08/2024] [Accepted: 08/16/2024] [Indexed: 08/26/2024]
Abstract
The global attention on microplastic pollution and its implications for human health has grown in recent years. Additionally, the co-existence of heavy metals may significantly alter microplastics' physicochemical characteristics, potentially amplifying their overall toxicity-a facet that remains less understood. In this study, we focused the membrane toxicity of modified polystyrene microplastics (PS-MPs) following cadmium (Cd) pretreatment. Our findings revealed that Cd-pretreated PS-MPs exacerbated their toxic effects, including diminished membrane integrity and altered phase fluidity in simulated lipid membrane giant unilamellar vesicles (GUVs), as well as heightened membrane permeability, protein damage, and lipid peroxidation in red blood cells and macrophages. Mechanistically, these augmented membrane toxicities can be partially ascribed to modifications in the surface roughness and hydrophilicity of Cd-pretreated PS-MPs, as well as to interactions between PS-MPs and lipid bilayers. Notably, hydrogen bonds emerged as a crucial mechanism underlying the enhanced interaction of PS-MPs with lipid bilayers.
Collapse
Affiliation(s)
- Wanqing Zhao
- College of Environmental Science and Engineering, Hebei University of Science and Technology, Hebei Key Laboratory of Pollution Prevention Biotechnology, Shijiazhuang 050018, China
| | - Tong Ye
- College of Environmental Science and Engineering, Hebei University of Science and Technology, Hebei Key Laboratory of Pollution Prevention Biotechnology, Shijiazhuang 050018, China
| | - Jianwen Zhou
- College of Environmental Science and Engineering, Hebei University of Science and Technology, Hebei Key Laboratory of Pollution Prevention Biotechnology, Shijiazhuang 050018, China
| | - Xuan Zhang
- College of Environmental Science and Engineering, Hebei University of Science and Technology, Hebei Key Laboratory of Pollution Prevention Biotechnology, Shijiazhuang 050018, China; Medical Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250062, China
| | - Ke Wang
- Hebei Key Laboratory of Intractable Pathogens, Shijiazhuang Center for Disease Control and Prevention, Shijiazhuang 050011, China
| | - Hong Zhang
- Hebei Key Laboratory of Intractable Pathogens, Shijiazhuang Center for Disease Control and Prevention, Shijiazhuang 050011, China
| | - Jiansheng Cui
- College of Environmental Science and Engineering, Hebei University of Science and Technology, Hebei Key Laboratory of Pollution Prevention Biotechnology, Shijiazhuang 050018, China
| | - Shuping Zhang
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University, Jinan 250117, China; Medical Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250062, China.
| | - Lixin Wang
- College of Environmental Science and Engineering, Hebei University of Science and Technology, Hebei Key Laboratory of Pollution Prevention Biotechnology, Shijiazhuang 050018, China.
| |
Collapse
|
7
|
Lou Z, Mu C, Corpstein CD, Li T. In vivo deposition of poorly soluble drugs. Adv Drug Deliv Rev 2024; 211:115358. [PMID: 38851590 DOI: 10.1016/j.addr.2024.115358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 05/12/2024] [Accepted: 06/05/2024] [Indexed: 06/10/2024]
Abstract
Administered drug molecules, whether dissolved or solubilized, have the potential to precipitate and accumulate as solid forms in tissues and cells within the body. This phase transition can significantly impact the pharmacokinetics of treatment. It is thus crucial to gain an understanding of how drug solubility/permeability, drug formulations and routes of administration affect in vivo behaviors of drug deposition. This review examines literature reports on the drug deposition in tissues and cells of poorly water-soluble drugs, as well as underlying physical mechanisms that lead to precipitation. Our work particularly highlights drug deposition in macrophages and the subcellular fate of precipitated drugs. We also propose a tissue permeability-based classification framework to evaluate precipitation potentials of poorly soluble drugs in major organs and tissues. The impact on pharmacokinetics is further discussed and needs to be considered in developing drug delivery systems. Finally, bioimaging techniques that are used to examine aggregated states and the intracellular trafficking of absorbed drugs are summarized.
Collapse
Affiliation(s)
- Zhaohuan Lou
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Zhejiang, Hangzhou 310053, China; Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47906, USA
| | - Chaofeng Mu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Zhejiang, Hangzhou 310053, China
| | - Clairissa D Corpstein
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47906, USA
| | - Tonglei Li
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47906, USA.
| |
Collapse
|
8
|
Zhu Y, Choi D, Somanath PR, Zhang D. Lipid-Laden Macrophages in Pulmonary Diseases. Cells 2024; 13:889. [PMID: 38891022 PMCID: PMC11171561 DOI: 10.3390/cells13110889] [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: 04/21/2024] [Revised: 05/17/2024] [Accepted: 05/21/2024] [Indexed: 06/20/2024] Open
Abstract
Pulmonary surfactants play a crucial role in managing lung lipid metabolism, and dysregulation of this process is evident in various lung diseases. Alternations in lipid metabolism lead to pulmonary surfactant damage, resulting in hyperlipidemia in response to lung injury. Lung macrophages are responsible for recycling damaged lipid droplets to maintain lipid homeostasis. The inflammatory response triggered by external stimuli such as cigarette smoke, bleomycin, and bacteria can interfere with this process, resulting in the formation of lipid-laden macrophages (LLMs), also known as foamy macrophages. Recent studies have highlighted the potential significance of LLM formation in a range of pulmonary diseases. Furthermore, growing evidence suggests that LLMs are present in patients suffering from various pulmonary conditions. In this review, we summarize the essential metabolic and signaling pathways driving the LLM formation in chronic obstructive pulmonary disease, pulmonary fibrosis, tuberculosis, and acute lung injury.
Collapse
Affiliation(s)
- Yin Zhu
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA 30912, USA (D.C.)
- Charlie Norwood VA Medical Center, Augusta, GA 30912, USA
| | - Dooyoung Choi
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA 30912, USA (D.C.)
| | - Payaningal R. Somanath
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA 30912, USA (D.C.)
- Charlie Norwood VA Medical Center, Augusta, GA 30912, USA
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Duo Zhang
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA 30912, USA (D.C.)
- Charlie Norwood VA Medical Center, Augusta, GA 30912, USA
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| |
Collapse
|
9
|
Lv S, Cao M, Luo J, Fu K, Yuan W. Search progress of pyruvate kinase M2 (PKM2) in organ fibrosis. Mol Biol Rep 2024; 51:389. [PMID: 38446272 DOI: 10.1007/s11033-024-09307-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 02/01/2024] [Indexed: 03/07/2024]
Abstract
Fibrosis is characterized by abnormal deposition of the extracellular matrix (ECM), leading to organ structural remodeling and loss of function. The principal cellular effector in fibrosis is activated myofibroblasts, which serve as the main source of matrix proteins. Metabolic reprogramming, transitioning from mitochondrial oxidative phosphorylation to aerobic glycolysis, is widely observed in rapidly dividing cells such as tumor cells and activated myofibroblasts and is increasingly recognized as a fundamental pathogenic basis in organ fibrosis. Targeting metabolism represents a promising strategy to mitigate fibrosis. PKM2, a key enzyme in glycolysis, plays a pivotal role in metabolic reprogramming through allosteric regulation, impacting both metabolic and non-metabolic pathways. Therefore, metabolic reprogramming induced by PKM2 activation is involved in the occurrence and development of fibrosis in various organs. A comprehensive understanding of the role of PKM2 in fibrotic diseases is crucial for seeking new anti-fibrotic therapeutic targets. In this context, we summarize PKM2's role in glycolysis, mediating the intricate mechanisms underlying fibrosis in multiple organs, and discuss the potential value of PKM2 inhibitors and allosteric activators in future clinical treatments, aiming to identify novel therapeutic targets for proliferative fibrotic diseases.
Collapse
Affiliation(s)
- Shumei Lv
- Department of Cardiology, Hospital of Jiangsu University, Zhenjiang, Jiangsu, 212000, China
| | - Mengfei Cao
- Department of Cardiology, Hospital of Jiangsu University, Zhenjiang, Jiangsu, 212000, China
| | - Jie Luo
- Department of Cardiology, Hospital of Jiangsu University, Zhenjiang, Jiangsu, 212000, China
| | - Kewei Fu
- Department of Cardiology, Hospital of Jiangsu University, Zhenjiang, Jiangsu, 212000, China
| | - Wei Yuan
- Department of Cardiology, Hospital of Jiangsu University, Zhenjiang, Jiangsu, 212000, China.
| |
Collapse
|
10
|
Bahiraii S, Brenner M, Weckwerth W, Heiss EH. Sulforaphane impedes mitochondrial reprogramming and histone acetylation in polarizing M1 (LPS) macrophages. Free Radic Biol Med 2024; 213:443-456. [PMID: 38301976 DOI: 10.1016/j.freeradbiomed.2024.01.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 01/18/2024] [Indexed: 02/03/2024]
Abstract
M1 (LPS) macrophages are characterized by a high expression of pro-inflammatory mediators, and distinct metabolic features that comprise increased glycolysis, a broken TCA cycle, or impaired OXPHOS with augmented mitochondrial ROS production. This study investigated whether the phytochemical sulforaphane (Sfn) influences mitochondrial reprogramming during M1 polarization, as well as to what extent this can contribute to Sfn-mediated inhibition of M1 marker expression in murine macrophages. The use of extracellular flux-, metabolite-, and immunoblot analyses as well as fluorescent dyes indicative for mitochondrial morphology, membrane potential or superoxide production, demonstrated that M1 (LPS/Sfn) macrophages maintain an unbroken TCA cycle, higher OXPHOS rate, boosted fusion dynamics, lower membrane potential, and less superoxide production in their mitochondria when compared to control M1 (LPS) cells. Sustained OXPHOS and TCA activity but not the concomitantly observed high dependency on fatty acids as fuel appeared necessary for M1 (LPS/Sfn) macrophages to reduce expression of nos2, il1β, il6 and tnfα. M1 (LPS/Sfn) macrophages also displayed lower nucleo/cytosolic acetyl-CoA levels in association with lower global and site-specific histone acetylation at selected pro-inflammatory gene promoters than M1 (LPS), evident in colorimetric coupled enzyme assays, immunoblot and ChIP-qPCR analyses, respectively. Supplementation with acetate or citrate was able to rescue both histone acetylation and mRNA expression of the investigated M1 marker genes in Sfn-treated cells. Overall, Sfn preserves mitochondrial functionality and restricts indispensable nuclear acetyl-CoA for histone acetylation and M1 marker expression in LPS-stimulated macrophages.
Collapse
Affiliation(s)
- Sheyda Bahiraii
- Department of Pharmaceutical Sciences, University of Vienna, Vienna, Austria; ViennaDoctoral School of Pharmaceutical, Nutritional and Sport Sciences (VDS PhaNuSpo), University of Vienna, Vienna, Austria
| | - Martin Brenner
- Department of Pharmaceutical Sciences, University of Vienna, Vienna, Austria; ViennaDoctoral School of Pharmaceutical, Nutritional and Sport Sciences (VDS PhaNuSpo), University of Vienna, Vienna, Austria; Vienna Metabolomics Center (VIME), University of Vienna, Vienna, Austria
| | - Wolfram Weckwerth
- Vienna Metabolomics Center (VIME), University of Vienna, Vienna, Austria; Molecular Systems Biology (MOSYS), Department of Functional and Evolutionary Ecology (FEE), University of Vienna, Vienna, Austria
| | - Elke H Heiss
- Department of Pharmaceutical Sciences, University of Vienna, Vienna, Austria.
| |
Collapse
|
11
|
Yang C, Xue Y, Duan Y, Mao C, Wan M. Extracellular vesicles and their engineering strategies, delivery systems, and biomedical applications. J Control Release 2024; 365:1089-1123. [PMID: 38065416 DOI: 10.1016/j.jconrel.2023.11.057] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 11/27/2023] [Accepted: 11/29/2023] [Indexed: 01/07/2024]
Abstract
Extracellular vesicles are nanoscale vesicles that can be secreted by all cell types, are intracellular in origin and have the same composition as their parent cells, play a key role in intercellular communication in organismal health and disease, and are now often used as biomarkers of disease and therapeutic agents in biomedical research. When injected locally or systemically, they have the ability to provide a variety of therapeutic effects, for example, regeneration of skin damage or restoration of cardiac function. However, direct injection of extracellular vesicles may result in their rapid clearance from the injection site.In order to maintain the biological activity of extracellular vesicles and to control the release of effective concentrations for better therapeutic efficacy during long-term disease treatment, the design of an optimized drug delivery system is necessary and different systems for the continuous delivery of extracellular vesicles have been developed. This paper first provides an overview of the biogenesis, composition and physiological function of extracellular vesicles, followed by a review of different strategies for extracellular vesicle isolation and methods for engineering extracellular vesicles. In addition, this paper reviews the latest extracellular vesicle delivery platforms such as micro-nanoparticles, injectable hydrogels, microneedles and scaffold patches. At the same time, the research progress and key cases of extracellular vesicle delivery systems in the field of biomedical therapeutics are described. Finally, the challenges and future trends of extracellular vesicle delivery are discussed.
Collapse
Affiliation(s)
- Chunhao Yang
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Yunxin Xue
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Yu Duan
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Chun Mao
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China.
| | - Mimi Wan
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China.
| |
Collapse
|
12
|
Stevenson ER, Smith LC, Wilkinson ML, Lee SJ, Gow AJ. Etiology of lipid-laden macrophages in the lung. Int Immunopharmacol 2023; 123:110719. [PMID: 37595492 PMCID: PMC10734282 DOI: 10.1016/j.intimp.2023.110719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 07/18/2023] [Accepted: 07/25/2023] [Indexed: 08/20/2023]
Abstract
Uniquely positioned as sentinel cells constantly exposed to the environment, pulmonary macrophages are vital for the maintenance of the lung lining. These cells are responsible for the clearance of xenobiotics, pathogen detection and clearance, and homeostatic functions such as surfactant recycling. Among the spectrum of phenotypes that may be expressed by macrophages in the lung, the pulmonary lipid-laden phenotype is less commonly studied in comparison to its circulatory counterpart, the atherosclerotic lesion-associated foam cell, or the acutely activated inflammatory macrophage. Herein, we propose that lipid-laden macrophage formation in the lung is governed by lipid acquisition, storage, metabolism, and export processes. The cellular balance of these four processes is critical to the maintenance of homeostasis and the prevention of aberrant signaling that may contribute to lung pathologies. This review aims to examine mechanisms and signaling pathways that are involved in lipid-laden macrophage formation and the potential consequences of this phenotype in the lung.
Collapse
Affiliation(s)
- E R Stevenson
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ, United States
| | - L C Smith
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ, United States; Department of Pharmaceutical Sciences, University of Connecticut School of Pharmacy, Storrs, CT, United States
| | - M L Wilkinson
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ, United States
| | - S J Lee
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ, United States
| | - A J Gow
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ, United States
| |
Collapse
|
13
|
Hu L, Ter Hofstede B, Sharma D, Zhao F, Walsh AJ. Comparison of phasor analysis and biexponential decay curve fitting of autofluorescence lifetime imaging data for machine learning prediction of cellular phenotypes. FRONTIERS IN BIOINFORMATICS 2023; 3:1210157. [PMID: 37455808 PMCID: PMC10342207 DOI: 10.3389/fbinf.2023.1210157] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 06/21/2023] [Indexed: 07/18/2023] Open
Abstract
Introduction: Autofluorescence imaging of the coenzymes reduced nicotinamide (phosphate) dinucleotide (NAD(P)H) and oxidized flavin adenine dinucleotide (FAD) provides a label-free method to detect cellular metabolism and phenotypes. Time-domain fluorescence lifetime data can be analyzed by exponential decay fitting to extract fluorescence lifetimes or by a fit-free phasor transformation to compute phasor coordinates. Methods: Here, fluorescence lifetime data analysis by biexponential decay curve fitting is compared with phasor coordinate analysis as input data to machine learning models to predict cell phenotypes. Glycolysis and oxidative phosphorylation of MCF7 breast cancer cells were chemically inhibited with 2-deoxy-d-glucose and sodium cyanide, respectively; and fluorescence lifetime images of NAD(P)H and FAD were obtained using a multiphoton microscope. Results: Machine learning algorithms built from either the extracted lifetime values or phasor coordinates predict MCF7 metabolism with a high accuracy (∼88%). Similarly, fluorescence lifetime images of M0, M1, and M2 macrophages were acquired and analyzed by decay fitting and phasor analysis. Machine learning models trained with features from curve fitting discriminate different macrophage phenotypes with improved performance over models trained using only phasor coordinates. Discussion: Altogether, the results demonstrate that both curve fitting and phasor analysis of autofluorescence lifetime images can be used in machine learning models for classification of cell phenotype from the lifetime data.
Collapse
Affiliation(s)
| | | | | | | | - Alex J. Walsh
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, United States
| |
Collapse
|
14
|
Li S, Wang C, Tang YD, Qin L, Chen T, Wang S, Bai Y, Cai X, Wang S. Interaction between Porcine Alveolar Macrophage-Tang Cells and Streptococcus suis Strains of Different Virulence: Phagocytosis and Apoptosis. Microorganisms 2023; 11:microorganisms11010160. [PMID: 36677452 PMCID: PMC9863715 DOI: 10.3390/microorganisms11010160] [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: 11/24/2022] [Revised: 12/21/2022] [Accepted: 01/06/2023] [Indexed: 01/11/2023] Open
Abstract
Streptococcus suis is an important swine bacterial pathogen that activates macrophages to secrete inflammatory cytokines. Primary porcine alveolar macrophages (PAMs) are inconvenient to obtain, but it is unknown whether immortalized PAM-Tang cells can replace them as a better cell model for the study of the interaction between S. suis and macrophages. In this study, the phagocytic integrity, polarization, and pro-inflammatory cytokine secretion of PAM-Tang cells were confirmed by live-cell imaging, electron microscopy, confocal microscopy, and ELISA. Interestingly, the S. suis serotype 9 avirulent strain W7119 induced higher levels of adhesion and pro-inflammatory cytokines in PAM-Tang cells than the S. suis serotype 2 virulent strain 700794. Prolonged incubation with S. suis caused more cytotoxic cell damage, and the virulent strain induced higher levels of cytotoxicity to PAM-Tang cells. The virulent strain also induced higher levels of apoptosis in PAM-Tang cells, as shown by terminal deoxynucleotidyl transferase (TdT)-mediated dUTP-biotin nick end labeling (TUNEL) assay. In addition, it is the first report of virulent and avirulent S. suis inducing PAM-Tang polarization towards pro-inflammatory M1 macrophages and p53- and caspase-dependent apoptosis in PAMs. Taken together, this study contributes to a better understand of interactions between macrophages and S. suis isolates of different virulence, and confirms that PAM-Tang cells provide a long-term, renewable resource for investigating macrophage infections with bacteria.
Collapse
Affiliation(s)
- Siqi Li
- National Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, China
| | - Chunsheng Wang
- College of Life Science, Northeast Forestry University, Harbin 150040, China
| | - Yan-Dong Tang
- National Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, China
| | - Lei Qin
- National Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, China
| | - Tianfeng Chen
- National Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, China
| | - Shanghui Wang
- National Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, China
| | - Yuanzhe Bai
- National Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, China
| | - Xuehui Cai
- National Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, China
- Correspondence: (X.C.); (S.W.)
| | - Shujie Wang
- National Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, China
- Heilongjiang Provincial Key Laboratory of Veterinary Immunology, Harbin 150069, China
- Correspondence: (X.C.); (S.W.)
| |
Collapse
|
15
|
Lin J, Jiang L, Guo K, Feng N. Decreased VEGFA alleviates the symptoms of LPS-induced sepsis in a mouse model by inhibiting glycolysis and thereby regulating the polarization of macrophages. Eur J Histochem 2022; 67:3528. [PMID: 36546420 PMCID: PMC9827425 DOI: 10.4081/ejh.2023.3528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 12/01/2022] [Indexed: 12/24/2022] Open
Abstract
The immune imbalance caused by excessive inflammatory reactions is the primary cause of sepsis. Macrophages with M1 and M2 polarization states are important immune cells that regulate the balance of the inflammatory response in sepsis. Encouraging the conversion of macrophages from the M1 to the M2 type is an important strategy for relieving sepsis. Here, we demonstrated the upregulation of vascular endothelial growth factor A (VEGFA) in a mouse model of sepsis. Then, siRNA technology was applied to inhibit the expression of VEGFA in macrophages. Flow cytometry and RT‒qPCR results showed that low expression of VEGFA inhibited LPS-induced M1 polarization of macrophages. Decreased VEGFA was also proven to lower TNF-α, IL-1β, and IL-6 secretion by LPS-induced macrophages. In addition, the effects of knocking down VEGFA on the energy metabolism pattern of macrophages were investigated by glycolysis pressure tests and mitochondrial pressure tests, and VEGFA knockdown reversed the induction of glycolysis in macrophages by LPS. The mitochondrial content and ATP content results also confirmed this finding. After the tail vein of septic mice was injected with macrophages transfected with si-VEGFA, the liver and kidney damage and the pathological conditions of the lung were alleviated. The secretion of TNF-α and IL-6 was decreased, while IL-10 was increased in their serum. Immunohistochemical staining revealed decreased expression of CD86 and increased expression of CD206 in the si-VEGFA group. This study demonstrates that decreased VEGFA inhibits glycolysis and thus inhibits LPS-induced M1 polarization of macrophages, ultimately relieving sepsis.
Collapse
Affiliation(s)
- Jun Lin
- Intensive Care Unit, Pingxiang People’s Hospital, Jiangxi
| | - Liping Jiang
- Division of Science and Education, Pingxiang People’s Hospital, Jiangxi, China
| | - Kun Guo
- Intensive Care Unit, Pingxiang People’s Hospital, Jiangxi
| | - Ning Feng
- Intensive Care Unit, Pingxiang People’s Hospital, Jiangxi,Intensive Care Unit, Pingxiang People’s Hospital. No. 8, Zhongshan Avenue, Pingxiang, Jiangxi 337055, China.
| |
Collapse
|
16
|
Boroumand P, Prescott DC, Mukherjee T, Bilan PJ, Wong M, Shen J, Tattoli I, Zhou Y, Li A, Sivasubramaniyam T, Shi N, Zhu LY, Liu Z, Robbins C, Philpott DJ, Girardin SE, Klip A. Bone marrow adipocytes drive the development of tissue invasive Ly6C high monocytes during obesity. eLife 2022; 11:65553. [PMID: 36125130 PMCID: PMC9512398 DOI: 10.7554/elife.65553] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 09/06/2022] [Indexed: 11/13/2022] Open
Abstract
During obesity and high fat-diet (HFD) feeding in mice, sustained low-grade inflammation includes not only increased pro-inflammatory macrophages in the expanding adipose tissue, but also bone marrow (BM) production of invasive Ly6Chigh monocytes. As BM adiposity also accrues with HFD, we explored the relationship between the gains in BM white adipocytes and invasive Ly6Chigh monocytes by in vivo and ex vivo paradigms. We find a temporal and causal link between BM adipocyte whitening and the Ly6Chigh monocyte surge, preceding the adipose tissue macrophage rise during HFD in mice. Phenocopying this, ex vivo treatment of BM cells with conditioned media from BM adipocytes or bona fide white adipocytes favoured Ly6Chigh monocyte preponderance. Notably, Ly6Chigh skewing was preceded by monocyte metabolic reprogramming towards glycolysis, reduced oxidative potential and increased mitochondrial fission. In sum, short-term HFD changes BM cellularity, resulting in local adipocyte whitening driving a gradual increase and activation of invasive Ly6Chigh monocytes.
Collapse
Affiliation(s)
| | - David C Prescott
- Department of Immunology, University of Toronto, Toronto, Canada
| | - Tapas Mukherjee
- Department of Immunology, University of Toronto, Toronto, Canada
| | - Philip J Bilan
- Cell Biology Program, Hospital for Sick Children, Toronto, Canada
| | - Michael Wong
- Cell Biology Program, Hospital for Sick Children, Toronto, Canada
| | - Jeff Shen
- Cell Biology Program, Hospital for Sick Children, Toronto, Canada
| | - Ivan Tattoli
- Department of Laboratory Medicine and Pathopysiology, University of Toronto, Toronto, Canada
| | - Yuhuan Zhou
- Cell Biology Program, Hospital for Sick Children, Toronto, Canada
| | - Angela Li
- Research Institute, Toronto General Hospital, Toronto, Canada
| | | | - Nan Shi
- Cell Biology Program, Hospital for Sick Children, Toronto, Canada
| | - Lucie Y Zhu
- Cell Biology Program, Hospital for Sick Children, Toronto, Canada
| | - Zhi Liu
- Cell Biology Program, Hospital for Sick Children, Toronto, Canada
| | - Clinton Robbins
- Department of Laboratory Medicine and Pathophysiology, University of Toronto, Toronto, Canada
| | - Dana J Philpott
- Department of Immunology, University of Toronto, Toronto, Canada
| | | | - Amira Klip
- Cell Biology Program, Hospital for Sick Children, Toronto, Canada
| |
Collapse
|
17
|
Sun JX, Xu XH, Jin L. Effects of Metabolism on Macrophage Polarization Under Different Disease Backgrounds. Front Immunol 2022; 13:880286. [PMID: 35911719 PMCID: PMC9331907 DOI: 10.3389/fimmu.2022.880286] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 06/21/2022] [Indexed: 11/20/2022] Open
Abstract
Macrophages are versatile immune cells associated with various diseases, and their phenotypes and functions change on the basis of the surrounding environments. Reprogramming of metabolism is required for the proper polarization of macrophages. This review will focus on basic metabolic pathways, the effects of key enzymes and specific products, relationships between cellular metabolism and macrophage polarization in different diseases and the potential prospect of therapy targeted key metabolic enzymes. In particular, the types and characteristics of macrophages at the maternal-fetal interface and their effects on a successful conception will be discussed.
Collapse
Affiliation(s)
| | | | - Liping Jin
- *Correspondence: Liping Jin, ; Xiang-Hong Xu,
| |
Collapse
|
18
|
O’Mahony C, Amamou A, Ghosh S. Diet-Microbiota Interplay: An Emerging Player in Macrophage Plasticity and Intestinal Health. Int J Mol Sci 2022; 23:3901. [PMID: 35409260 PMCID: PMC8998881 DOI: 10.3390/ijms23073901] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/28/2022] [Accepted: 03/30/2022] [Indexed: 02/04/2023] Open
Abstract
Inflammatory bowel diseases (IBD) are chronic disorders of the gastrointestinal tract with an increasing prevalence worldwide. Targeted therapies for IBD are limited by several factors, including the therapeutic ceiling and the high incidence of non-responders or loss-of-response. In order to improve therapeutic efficacy, there is critical need to decipher disease pathogenesis, currently not well understood. Macrophages, innate immune cells that exhibit high plasticity, perpetuate inflammatory signalling in IBD through excessive release of inflammatory mediators. In recent years, pioneering research has revealed the importance of the interplay between macrophages and gut microbiota in maintaining intestinal homeostasis. Particular attention is focusing on microbiota-derived metabolites, believed to possess immunomodulatory properties capable of manipulating macrophage plasticity. Microbiota-derived short-chain fatty acids (SCFAs) and indole compounds, along with dietary sourced omega-3 (ω-3) polyunsaturated fatty acids (PUFA), exert anti-inflammatory effects, attributable to interactions with macrophages. Before we can effectively incorporate these metabolites into IBD therapies, a deeper understanding of microbiota-macrophage interactions at a molecular level is necessary. Therefore, the aim of this review is firstly to detail current knowledge regarding how diet and microbiota-derived metabolites modify macrophage plasticity. Later, we discuss the concept of therapeutic strategies directed at microbiota-macrophage interactions, which could be highly valuable for IBD therapies in the future.
Collapse
Affiliation(s)
- Cian O’Mahony
- APC Microbiome Ireland, College of Medicine and Health, University College Cork, T12 YT20 Cork, Ireland; (A.A.); (S.G.)
| | | | | |
Collapse
|
19
|
Kosanović M, Milutinovic B, Glamočlija S, Morlans IM, Ortiz A, Bozic M. Extracellular Vesicles and Acute Kidney Injury: Potential Therapeutic Avenue for Renal Repair and Regeneration. Int J Mol Sci 2022; 23:ijms23073792. [PMID: 35409151 PMCID: PMC8998560 DOI: 10.3390/ijms23073792] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 03/27/2022] [Accepted: 03/28/2022] [Indexed: 12/10/2022] Open
Abstract
Acute kidney injury (AKI) is a sudden decline of renal function and represents a global clinical problem due to an elevated morbidity and mortality. Despite many efforts, currently there are no treatments to halt this devastating condition. Extracellular vesicles (EVs) are nanoparticles secreted by various cell types in both physiological and pathological conditions. EVs can arise from distinct parts of the kidney and can mediate intercellular communication between various cell types along the nephron. Besides their potential as diagnostic tools, EVs have been proposed as powerful new tools for regenerative medicine and have been broadly studied as therapeutic mediators in different models of experimental AKI. In this review, we present an overview of the basic features and biological relevance of EVs, with an emphasis on their functional role in cell-to-cell communication in the kidney. We explore versatile roles of EVs in crucial pathophysiological mechanisms contributing to AKI and give a detailed description of the renoprotective effects of EVs from different origins in AKI. Finally, we explain known mechanisms of action of EVs in AKI and provide an outlook on the potential clinical translation of EVs in the setting of AKI.
Collapse
Affiliation(s)
- Maja Kosanović
- Institute for the Application of Nuclear Energy, INEP, University of Belgrade, 11080 Belgrade, Serbia; (M.K.); (S.G.)
| | - Bojana Milutinovic
- Department of Neurosurgery, MD Anderson Cancer Center, University of Texas, Houston, TX 77030, USA;
| | - Sofija Glamočlija
- Institute for the Application of Nuclear Energy, INEP, University of Belgrade, 11080 Belgrade, Serbia; (M.K.); (S.G.)
| | - Ingrid Mena Morlans
- Vascular and Renal Translational Research Group, Biomedical Research Institute of Lleida Dr. Pifarré Foundation (IRBLleida), 25196 Lleida, Spain;
| | - Alberto Ortiz
- Department of Nephrology and Hypertension, IIS-Fundación Jiménez Díaz, Universidad Autónoma Madrid, 28040 Madrid, Spain;
| | - Milica Bozic
- Vascular and Renal Translational Research Group, Biomedical Research Institute of Lleida Dr. Pifarré Foundation (IRBLleida), 25196 Lleida, Spain;
- Correspondence:
| |
Collapse
|
20
|
Li N, Chen J, Geng C, Wang X, Wang Y, Sun N, Wang P, Han L, Li Z, Fan H, Hou S, Gong Y. Myoglobin promotes macrophage polarization to M1 type and pyroptosis via the RIG-I/Caspase1/GSDMD signaling pathway in CS-AKI. Cell Death Dis 2022; 8:90. [PMID: 35228524 PMCID: PMC8885737 DOI: 10.1038/s41420-022-00894-w] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 01/29/2022] [Accepted: 02/11/2022] [Indexed: 12/14/2022]
Abstract
Crush syndrome (CS) is a life-threatening illness in traffic accidents and earthquakes. Crush syndrome-induced acute kidney injury (CS-AKI) is considered to be mainly due to myoglobin (Mb) circulation and deposition after skeletal muscle ruptures and releases. Macrophages are the primary immune cells that fight foreign substances and play critical roles in regulating the body's natural immune response. However, what effect does myoglobin have on macrophages and the mechanisms involved in the CS-AKI remain unclear. This study aims to look into how myoglobin affects macrophages of the CS-AKI model. C57BL/6 mice were used to construct the CS-AKI model by digital crush platform. Biochemical analysis and renal histology confirmed the successful establishment of the CS-AKI mouse model. Ferrous myoglobin was used to treat Raw264.7 macrophages to mimic the CS-AKI cell model in vitro. The macrophage polarization toward M1 type and activation of RIG-I as myoglobin sensor were verified by real-time quantitative PCR (qPCR), Western blotting (WB), and immunofluorescence (IF). Macrophage pyroptosis was observed under light microscopy. The interaction between RIG-I and caspase1 was subsequently explored by co-immunoprecipitation (Co-IP) and IF. Small interfering RNA (siRIG-I) and pyroptosis inhibitor dimethyl fumarate (DMF) were used to verify the role of macrophage polarization and pyroptosis in CS-AKI. In the kidney tissue of CS-AKI mice, macrophage infiltration and M1 type were found. We also detected that in the cell model of CS-AKI in vitro, ferrous myoglobin treatment promoted macrophages polarization to M1. Meanwhile, we observed pyroptosis, and myoglobin activated the RIG-I/Caspase1/GSDMD signaling pathway. In addition, pyroptosis inhibitor DMF not only alleviated kidney injury of CS-AKI mice but also inhibited macrophage polarization to M1 phenotype and pyroptosis via the RIG-I/Caspase1/GSDMD signaling pathway. Our research found that myoglobin promotes macrophage polarization to M1 type and pyroptosis via the RIG-I/Caspase1/GSDMD signaling pathway in CS-AKI.
Collapse
Affiliation(s)
- Ning Li
- Wenzhou Safety (Emergency) Institute, Tianjin University, Wenzhou, 325000, China.,Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, 300072, China.,State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300350, China.,Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin, 300072, China
| | - Jiale Chen
- Wenzhou Safety (Emergency) Institute, Tianjin University, Wenzhou, 325000, China.,Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, 300072, China.,Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin, 300072, China
| | - Chenhao Geng
- Wenzhou Safety (Emergency) Institute, Tianjin University, Wenzhou, 325000, China.,Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, 300072, China.,Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin, 300072, China
| | - Xinyue Wang
- Wenzhou Safety (Emergency) Institute, Tianjin University, Wenzhou, 325000, China.,Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, 300072, China.,Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin, 300072, China
| | - Yuru Wang
- Wenzhou Safety (Emergency) Institute, Tianjin University, Wenzhou, 325000, China.,Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, 300072, China.,Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin, 300072, China
| | - Na Sun
- Wenzhou Safety (Emergency) Institute, Tianjin University, Wenzhou, 325000, China.,Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, 300072, China.,Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin, 300072, China
| | - Pengtao Wang
- Department of Intensive Care Unit, Tianjin First Center Hospital, Tianjin, 300192, China
| | - Lu Han
- Wenzhou Safety (Emergency) Institute, Tianjin University, Wenzhou, 325000, China.,Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, 300072, China.,Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin, 300072, China
| | - Zizheng Li
- Wenzhou Safety (Emergency) Institute, Tianjin University, Wenzhou, 325000, China.,Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, 300072, China.,Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin, 300072, China
| | - Haojun Fan
- Wenzhou Safety (Emergency) Institute, Tianjin University, Wenzhou, 325000, China.,Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, 300072, China.,Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin, 300072, China
| | - Shike Hou
- Wenzhou Safety (Emergency) Institute, Tianjin University, Wenzhou, 325000, China. .,Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, 300072, China. .,Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin, 300072, China.
| | - Yanhua Gong
- Wenzhou Safety (Emergency) Institute, Tianjin University, Wenzhou, 325000, China. .,Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, 300072, China. .,Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin, 300072, China.
| |
Collapse
|
21
|
Li Y, Tan J, Miao Y, Zhang Q. MicroRNA in extracellular vesicles regulates inflammation through macrophages under hypoxia. Cell Death Dis 2021; 7:285. [PMID: 34635652 PMCID: PMC8505641 DOI: 10.1038/s41420-021-00670-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 08/25/2021] [Accepted: 09/14/2021] [Indexed: 12/24/2022]
Abstract
Extracellular vesicle (EV), critical mediators of cell-cell communication, allow cells to exchange proteins, lipids, and genetic material and therefore profoundly affect the general homeostasis. A hypoxic environment can affect the biogenesis and secrete of EVs, and the cargoes carried can participate in a variety of physiological and pathological processes. In hypoxia-induced inflammation, microRNA(miRNA) in EV participates in transcriptional regulation through various pathways to promote or reduce the inflammatory response. Meanwhile, as an important factor of immune response, the polarization of macrophages is closely linked to miRNAs, which will eventually affect the inflammatory state. In this review, we outline the possible molecular mechanism of EV changes under hypoxia, focusing on the signaling pathways of several microRNAs involved in inflammation regulation and describing the process and mechanism of EV-miRNAs regulating macrophage polarization in hypoxic diseases.
Collapse
Affiliation(s)
- Ye Li
- grid.412645.00000 0004 1757 9434Department of Geriatrics, Tianjin Geriatrics Institute, Tianjin Medical University General Hospital, 300052 Tianjin, China
| | - Jin Tan
- grid.412645.00000 0004 1757 9434Department of Geriatrics, Tianjin Geriatrics Institute, Tianjin Medical University General Hospital, 300052 Tianjin, China
| | - Yuyang Miao
- grid.265021.20000 0000 9792 1228Tianjin Medical University, 300052 Tianjin, China
| | - Qiang Zhang
- grid.412645.00000 0004 1757 9434Department of Geriatrics, Tianjin Geriatrics Institute, Tianjin Medical University General Hospital, 300052 Tianjin, China
| |
Collapse
|
22
|
Zhao F, Guo Z, Hou F, Fan W, Wu B, Qian Z. Magnoflorine Alleviates "M1" Polarized Macrophage-Induced Intervertebral Disc Degeneration Through Repressing the HMGB1/Myd88/NF-κB Pathway and NLRP3 Inflammasome. Front Pharmacol 2021; 12:701087. [PMID: 34366853 PMCID: PMC8343137 DOI: 10.3389/fphar.2021.701087] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 07/12/2021] [Indexed: 12/16/2022] Open
Abstract
Intervertebral disc degeneration (IDD) is related to the deterioration of nucleus pulposus (NP) cells due to hypertrophic differentiation and calcification. The imbalance of pro-inflammatory (M1 type) and anti-inflammatory (M2 type) macrophages contributes to maintaining tissue integrity. Here, we aimed to probe the effect of Magnoflorine (MAG) on NP cell apoptosis mediated by “M1” polarized macrophages. THP-1 cells were treated with lipopolysaccharide (LPS) to induce “M1” polarized macrophages. Under the treatment with increasing concentrations of MAG, the expression of pro-inflammatory cytokines (IL-1β, IL-6, TNF-α, IL-18), high mobility group box protein 1 (HMGB1), as well as myeloid differentiation factor 88 (MyD88), nuclear factor kappa B (NF-κB) and NOD-like receptor 3 (NLRP3) inflammasomes in THP-1 cells were determined. What’s more, human NP cells were treated with the conditioned medium (CM) from THP-1 cells. The NP cell viability and apoptosis were evaluated. Western blot (WB) was adopted to monitor the expression of apoptosis-related proteins (Bax, Caspase3, and Caspase9), catabolic enzymes (MMP-3, MMP-13, ADAMTS-4, and ADAMTS-5), and extracellular matrix (ECM) compositions (collagen II and aggrecan) in NP cells. As a result, LPS evidently promoted the expression of pro-inflammatory cytokines and HMGB1, the MyD88-NF-κB activation, and the NLRP3 inflammasome profile in THP-1 cells, while MAG obviously inhibited the "M1″ polarization of THP-1 cells. After treatment with “M1” polarized THP-1 cell CM, NP cell viability was decreased, while cell apoptosis, the pro-inflammatory cytokines, apoptosis-related proteins, and catabolic enzymes were distinctly up-regulated, and ECM compositions were reduced. After treatment with MAG, NP cell damages were dramatically eased. Furthermore, MAG dampened the HMGB1 expression and inactivated the MyD88/NF-κB pathway and NLRP3 inflammasome in NP cells. In conclusion, this study confirmed that MAG alleviates “M1” polarized macrophage-mediated NP cell damage by inactivating the HMGB1-MyD88-NF-κB pathway and NLRP3 inflammasome, which provides a new reference for IDD treatment.
Collapse
Affiliation(s)
- Feng Zhao
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou 215006, China.,Department of Orthopedics, The Second Hospital of Shanxi Medical University, Taiyuan 030001, China
| | - Zhenye Guo
- Department of Orthopedics, The Second Hospital of Shanxi Medical University, Taiyuan 030001, China
| | - Fushan Hou
- Department of Orthopedics, The Second Hospital of Shanxi Medical University, Taiyuan 030001, China
| | - Wei Fan
- Department of Orthopedics, The Second Hospital of Shanxi Medical University, Taiyuan 030001, China
| | - Binqiang Wu
- Department of Orthopedics, The Second Hospital of Shanxi Medical University, Taiyuan 030001, China
| | - Zhonglai Qian
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| |
Collapse
|
23
|
Shan X, Zhang C, Mai C, Hu X, Cheng N, Chen W, Peng D, Wang L, Ji Z, Xie Y. The Biogenesis, Biological Functions, and Applications of Macrophage-Derived Exosomes. Front Mol Biosci 2021; 8:715461. [PMID: 34368234 PMCID: PMC8333870 DOI: 10.3389/fmolb.2021.715461] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 07/09/2021] [Indexed: 12/11/2022] Open
Abstract
Macrophage-derived exosomes have been implicated on the modulation of inflammatory processes. Recent studies have shown that macrophage-derived exosomes contribute to the progression of many diseases such as cancer, atherosclerosis, diabetes and heart failure. This review describes the biogenesis of macrophage-derived exosomes and their biological functions in different diseases. In addition, the challenges facing the use of macrophage-derived exosomes as delivery tools for drugs, genes, and proteins in clinical applications are described. The application of macrophage-derived exosomes in the diagnosis and treatment of diseases is also discussed.
Collapse
Affiliation(s)
- Xiaoxiao Shan
- School of Pharmacy, Anhui Academy of Chinese Medicine, , Anhui University of Traditional Chinese MedicineHefei, China.,Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China.,Anhui Province Key Laboratory of Pharmaceutical Technology and Application, Hefei, China
| | - Caiyun Zhang
- School of Pharmacy, Anhui Academy of Chinese Medicine, , Anhui University of Traditional Chinese MedicineHefei, China.,Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China.,Anhui Province Key Laboratory of Pharmaceutical Technology and Application, Hefei, China
| | - Chutian Mai
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Avenida Wai Long, China
| | - Xuerui Hu
- School of Pharmacy, Anhui Academy of Chinese Medicine, , Anhui University of Traditional Chinese MedicineHefei, China.,Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China.,Anhui Province Key Laboratory of Pharmaceutical Technology and Application, Hefei, China
| | - Nuo Cheng
- School of Pharmacy, Anhui Academy of Chinese Medicine, , Anhui University of Traditional Chinese MedicineHefei, China.,Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China.,Anhui Province Key Laboratory of Pharmaceutical Technology and Application, Hefei, China
| | - Weidong Chen
- School of Pharmacy, Anhui Academy of Chinese Medicine, , Anhui University of Traditional Chinese MedicineHefei, China.,Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China.,Anhui Province Key Laboratory of Pharmaceutical Technology and Application, Hefei, China
| | - Daiyin Peng
- School of Pharmacy, Anhui Academy of Chinese Medicine, , Anhui University of Traditional Chinese MedicineHefei, China.,Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China
| | - Lei Wang
- School of Pharmacy, Anhui Academy of Chinese Medicine, , Anhui University of Traditional Chinese MedicineHefei, China.,Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China.,Anhui Province Key Laboratory of Pharmaceutical Technology and Application, Hefei, China
| | - Zhaojie Ji
- School of Pharmacy, Anhui Academy of Chinese Medicine, , Anhui University of Traditional Chinese MedicineHefei, China.,Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China
| | - Ying Xie
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Avenida Wai Long, China
| |
Collapse
|
24
|
Wang H, Yung MMH, Ngan HYS, Chan KKL, Chan DW. The Impact of the Tumor Microenvironment on Macrophage Polarization in Cancer Metastatic Progression. Int J Mol Sci 2021; 22:ijms22126560. [PMID: 34207286 PMCID: PMC8235734 DOI: 10.3390/ijms22126560] [Citation(s) in RCA: 138] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 06/07/2021] [Accepted: 06/14/2021] [Indexed: 02/07/2023] Open
Abstract
Rather than primary solid tumors, metastasis is one of the hallmarks of most cancer deaths. Metastasis is a multistage event in which cancer cells escape from the primary tumor survive in the circulation and disseminate to distant sites. According to Stephen Paget’s “Seed and Soil” hypothesis, metastatic capacity is determined not only by the internal oncogenic driving force but also by the external environment of tumor cells. Throughout the body, macrophages are required for maintaining tissue homeostasis, even in the tumor milieu. To fulfill these multiple functions, macrophages are polarized from the inflammation status (M1-like) to anti-inflammation status (M2-like) to maintain the balance between inflammation and regeneration. However, tumor cell-enforced tumor-associated macrophages (TAMs) (a high M2/M1 ratio status) are associated with poor prognosis for most solid tumors, such as ovarian cancer. In fact, clinical evidence has verified that TAMs, representing up to 50% of the tumor mass, exert both protumor and immunosuppressive effects in promoting tumor metastasis through secretion of interleukin 10 (IL10), transforming growth factor β (TGFβ), and VEGF, expression of PD-1 and consumption of arginine to inhibit T cell anti-tumor function. However, the underlying molecular mechanisms by which the tumor microenvironment favors reprogramming of macrophages to TAMs to establish a premetastatic niche remain controversial. In this review, we examine the latest investigations of TAMs during tumor development, the microenvironmental factors involved in macrophage polarization, and the mechanisms of TAM-mediated tumor metastasis. We hope to dissect the critical roles of TAMs in tumor metastasis, and the potential applications of TAM-targeted therapeutic strategies in cancer treatment are discussed.
Collapse
|
25
|
Li N, Chen J, Wang P, Fan H, Hou S, Gong Y. Major signaling pathways and key mediators of macrophages in acute kidney injury (Review). Mol Med Rep 2021; 23:455. [PMID: 33880578 PMCID: PMC8072315 DOI: 10.3892/mmr.2021.12094] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 03/17/2021] [Indexed: 12/16/2022] Open
Abstract
Acute kidney injury (AKI) has become a global public health problem with high morbidity and mortality rates, as well as high healthcare costs. Immune cells, particularly macrophages, which regulate tissue development, destroy pathogens, control homeostasis and repair wounds, play crucial and complex roles in AKI. In various types of AKI, numerous rapidly recruited monocytes and tissue-resident macrophages act in a coordinated manner. Thus, elucidating the phenotypic and functional characteristics of macrophages in AKI is essential for identifying potential therapeutic targets. Macrophage-sensing mediators and macrophage-derived mediators participate in the major macrophage-related signaling pathways in AKI, which regulate macrophage polarization and determine disease progression. In conclusion, macrophages change their roles and regulatory mechanisms during the occurrence and development of AKI. The aim of the present review was to contribute to an improved understanding of AKI and to the identification of novel therapeutic targets for this condition.
Collapse
Affiliation(s)
- Ning Li
- Institute of Disaster Medicine, Tianjin University, Tianjin 300072, P.R. China
| | - Jiale Chen
- Institute of Disaster Medicine, Tianjin University, Tianjin 300072, P.R. China
| | - Pengtao Wang
- Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin 300072, P.R. China
| | - Haojun Fan
- Institute of Disaster Medicine, Tianjin University, Tianjin 300072, P.R. China
| | - Shike Hou
- Institute of Disaster Medicine, Tianjin University, Tianjin 300072, P.R. China
| | - Yanhua Gong
- Institute of Disaster Medicine, Tianjin University, Tianjin 300072, P.R. China
| |
Collapse
|
26
|
Rehman A, Pacher P, Haskó G. Role of Macrophages in the Endocrine System. Trends Endocrinol Metab 2021; 32:238-256. [PMID: 33455863 DOI: 10.1016/j.tem.2020.12.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 12/07/2020] [Accepted: 12/08/2020] [Indexed: 12/19/2022]
Abstract
Macrophages are cells of the innate immune system that play myriad roles in the body. Macrophages are known to reside in endocrine glands, and a body of evidence now suggests that these cells interact closely with endocrine cells. Immune-endocrine interactions are important in the development of endocrine glands and their functioning during physiological states, and also become key players in pathophysiological states. Through gene expression profiling, diverse subpopulations of tissue macrophages have been discovered within endocrine organs; this has important implications for disease pathogenesis and potential pharmacotherapy. The molecular basis for the crosstalk between macrophages and endocrine cells is being unraveled, and allows the identification of multiple points for pharmacologic intervention. Macrophages in adipose tissue and pancreatic islets are key players in the process of metaflammation (metabolic inflammation) that underlies the development of insulin resistance, metabolic syndrome, diabetes mellitus, and non-alcoholic fatty liver disease. In the ovary, they play important roles in ovarian folliculogenesis and ovulation, whereas in the male reproductive tract they regulate spermatogenesis through the regulation of steroidogenesis by Leydig cells. We summarize the diverse roles played by macrophages in the endocrine system and identify potential targets for pharmacotherapy in endocrine disorders.
Collapse
Affiliation(s)
- Abdul Rehman
- Department of Medicine, Rutgers-New Jersey Medical School, Newark, NJ, USA
| | - Pál Pacher
- Laboratory of Cardiovascular Physiology and Tissue Injury, National Institutes of Health (NIH), National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, USA
| | - György Haskó
- Department of Anesthesiology, Columbia University, New York, NY, USA.
| |
Collapse
|
27
|
Guo C, Islam R, Zhang S, Fang J. Metabolic reprogramming of macrophages and its involvement in inflammatory diseases. EXCLI JOURNAL 2021; 20:628-641. [PMID: 33883988 PMCID: PMC8056050 DOI: 10.17179/excli2020-3053] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 03/03/2021] [Indexed: 12/28/2022]
Abstract
Macrophages are critical effector cells of the innate immune system. The presence of microbes or the stimulation by inflammatory factors triggers the metabolic reprogramming of macrophages or macrophage polarization into two phenotypes: the classically activated macrophages (M1) displaying a pro-inflammatory phenotype and the alternatively activated macrophages (M2) having anti-inflammatory functions. The imbalance between the two phenotypes has been linked with various pathological states, such as fibrosis, hepatitis, colitis, and tumor progression. An avenue of potential therapeutic strategies based on macrophage polarization has emerged. Therefore, it is essential to understand the mechanisms of macrophage polarization. In this review, we focus on the macrophage polarization process and discuss the stimuli-dependent conversion into M1 and M2 phenotypes. We also present the metabolic patterns supporting their specific functions. The factors and signaling cascades involved in intra-class switching are also detailed. Finally, the role of macrophage polarization in disease progression is discussed.
Collapse
Affiliation(s)
- Chunyu Guo
- Department of Toxicology, School of Public Health, Anhui Medical University, and Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, No 81 Meishan Road, Hefei 230032, China
| | - Rayhanul Islam
- Faculty of Pharmaceutical Science, Sojo University, Ikeda 4-22-1, Kumamoto 860-0082, Japan
| | - Shichen Zhang
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, China
| | - Jun Fang
- Department of Toxicology, School of Public Health, Anhui Medical University, and Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, No 81 Meishan Road, Hefei 230032, China.,Faculty of Pharmaceutical Science, Sojo University, Ikeda 4-22-1, Kumamoto 860-0082, Japan
| |
Collapse
|
28
|
Ying ZH, Li HM, Yu WY, Yu CH. Iridin Prevented Against Lipopolysaccharide-Induced Inflammatory Responses of Macrophages via Inactivation of PKM2-Mediated Glycolytic Pathways. J Inflamm Res 2021; 14:341-354. [PMID: 33574693 PMCID: PMC7872898 DOI: 10.2147/jir.s292244] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 01/12/2021] [Indexed: 12/27/2022] Open
Abstract
Purpose Abnormal glycolysis of immune cells contributed to the development of inflammatory response. Inhibition of this Warburg phenotype could be a promising strategy for preventing various inflammatory diseases. Iridin (IRD) is a natural isoflavone, and exerts anticancer, antioxidant, and anti-inflammatory effects. However, the underlying mechanism of IRD on acute inflammation remains unknown. In this study, the protective effects of IRD against lipopolysaccharide (LPS)-induced inflammation were investigated in murine macrophage RAW264.7 cells and in mice. Methods The inhibition of IRD on NO production in culture medium was detected by Griess assay while the levels of TNF-α, IL-1β, and MCP-1 were detected by ELISA assay. The effects of IRD on OCR and ECAR levels in LPS-treated macrophages were monitored by using Seahorse Analyzer. The apoptosis rate as well as the release of ROS and NO of RAW264.7 cells were analyzed by flow cytometric assay. The protective effects of IRD were investigated on LPS-induced inflammation in mice. The expressions of PKM2 and its downstream (p-JAK1, p-STAT1, p-STAT3, p-p65, iNOS, and COX2) in cells and in lung tissues were detected by Western blotting analysis. Results IRD treatment at the concentrations of 12.5-50 μM significantly inhibited the productions of TNF-α, IL-1β, MCP-1, and ROS, and suppressed the levels of glucose uptake and lactic acid in LPS-treated RAW264.7 cells. Oral administration with IRD (20-80 mg/kg) inhibited LPS-induced acute lung injury as well as inflammatory cytokine production in mice. Moreover, IRD targeted pyruvate kinase isozyme type M2 (PKM2) and suppressed its downstream p-JAK1, p-STAT1, p-STAT3, p-p65, iNOS, and COX2, which could be abolished by PKM2 agonist DASA-58 and antioxidant N-acetyl-L-cysteine, but partly be reversed by NF-κB activator CUT129 and JAK1 activator RO8191. Conclusion IRD alleviated LPS-induced inflammation through suppressing PKM2-mediated pathways, and could be a potential candidate for the prevention of inflammatory diseases.
Collapse
Affiliation(s)
- Zhen-Hua Ying
- Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, 310006, People's Republic of China
| | - Hui-Min Li
- Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, 310006, People's Republic of China
| | - Wen-Ying Yu
- Zhejiang Key Laboratory of Experimental Animal and Safety Evaluation, Hangzhou Medical College, Hangzhou, 310013, People's Republic of China
| | - Chen-Huan Yu
- Zhejiang Key Laboratory of Experimental Animal and Safety Evaluation, Hangzhou Medical College, Hangzhou, 310013, People's Republic of China.,Institute of Cancer and Basic Medicine, Chinese Academy of Sciences, Hangzhou, 310018, People's Republic of China.,Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, 310022, People's Republic of China
| |
Collapse
|
29
|
Mi R, Rabbi MH, Sun Y, Li X, Ma S, Wen Z, Meng N, Li Y, Du X, Li S. Enhanced protein phosphorylation in Apostichopus japonicus intestine triggered by tussah immunoreactive substances might be involved in the regulation of immune-related signaling pathways. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY D-GENOMICS & PROTEOMICS 2020; 37:100757. [PMID: 33197859 DOI: 10.1016/j.cbd.2020.100757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 10/25/2020] [Accepted: 10/29/2020] [Indexed: 11/28/2022]
Abstract
The sea cucumber Apostichopus japonicus is an economically important species owing to its high nutritive and medicinal value. In order to avoid the pollution resulting from the overuse of antibiotics in A. japonicus aquaculture, various immunostimulants have been used as an alternative to improve the efficiency of A. japonicus farming. Our previous proteomic investigation has shown that several proteins participating in the immune-related physiology of A. japonicus were differentially expressed in the intestinal tissue in response to tussah immunoreactive substances (TIS). This study further explored the immunostimulation mechanism of TIS in A. japonicus. Phosphoproteomics technology was used to investigate the effect of TIS on protein phosphorylation in the intestine of A. japonicus following feeding with a TIS-supplemented diet. A total of 213 unique phosphoproteins were detected from 225 unique phosphopeptides. KEGG pathway analysis showed that majority of the phosphoproteins are involved in endocytosis, carbon metabolism and spliceosome functional group. Sixteen of the phosphoproteins exhibited differential phosphorylation in response to TIS and 12 of these were found to associate with biological functions. Of these 12 phosphoproteins, eight exhibited enhanced phosphorylation while four displayed reduced phosphorylation. These 12 proteins were further analyzed and all were found to play a role in regulating some aspects of the immune system and the growth of sea cucumbers, especially in phagocytosis, energy metabolism and disease resistance. The findings of this study could therefore shed new light on the immune pathways of sea cucumber that are affected by TIS. This could help us to better understand the underlying mechanism linked to the immunoenhancement of A. japonicus in response to TIS, one that is associated with the change in protein phosphorylation.
Collapse
Affiliation(s)
- Rui Mi
- Liaoning Ocean and Fisheries Science Research Institute, Liaoning Academy of Agricultural Sciences, Dalian 116024, PR China
| | - Md Hasim Rabbi
- College of Fisheries and Life Science, Dalian Ocean University, Dalian 116024, PR China
| | - Yongxin Sun
- Liaoning Ocean and Fisheries Science Research Institute, Liaoning Academy of Agricultural Sciences, Dalian 116024, PR China.
| | - Xuejun Li
- Liaoning Ocean and Fisheries Science Research Institute, Liaoning Academy of Agricultural Sciences, Dalian 116024, PR China
| | - Shuhui Ma
- Liaoning Ocean and Fisheries Science Research Institute, Liaoning Academy of Agricultural Sciences, Dalian 116024, PR China
| | - Zhixin Wen
- Liaoning Ocean and Fisheries Science Research Institute, Liaoning Academy of Agricultural Sciences, Dalian 116024, PR China
| | - Nan Meng
- Liaoning Ocean and Fisheries Science Research Institute, Liaoning Academy of Agricultural Sciences, Dalian 116024, PR China
| | - Yajie Li
- Liaoning Ocean and Fisheries Science Research Institute, Liaoning Academy of Agricultural Sciences, Dalian 116024, PR China
| | - Xingfan Du
- Liaoning Ocean and Fisheries Science Research Institute, Liaoning Academy of Agricultural Sciences, Dalian 116024, PR China
| | - Shuying Li
- Liaoning Ocean and Fisheries Science Research Institute, Liaoning Academy of Agricultural Sciences, Dalian 116024, PR China
| |
Collapse
|
30
|
Heaster TM, Humayun M, Yu J, Beebe DJ, Skala MC. Autofluorescence Imaging of 3D Tumor-Macrophage Microscale Cultures Resolves Spatial and Temporal Dynamics of Macrophage Metabolism. Cancer Res 2020; 80:5408-5423. [PMID: 33093167 DOI: 10.1158/0008-5472.can-20-0831] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 07/17/2020] [Accepted: 10/19/2020] [Indexed: 12/24/2022]
Abstract
Macrophages within the tumor microenvironment (TME) exhibit a spectrum of protumor and antitumor functions, yet it is unclear how the TME regulates this macrophage heterogeneity. Standard methods to measure macrophage heterogeneity require destructive processing, limiting spatiotemporal studies of function within the live, intact 3D TME. Here, we demonstrate two-photon autofluorescence imaging of NAD(P)H and FAD to nondestructively resolve spatiotemporal metabolic heterogeneity of individual macrophages within 3D microscale TME models. Fluorescence lifetimes and intensities of NAD(P)H and FAD were acquired at 24, 48, and 72 hours poststimulation for mouse macrophages (RAW264.7) stimulated with IFNγ or IL4 plus IL13 in 2D culture, confirming that autofluorescence measurements capture known metabolic phenotypes. To quantify metabolic dynamics of macrophages within the TME, mouse macrophages or human monocytes (RAW264.7 or THP-1) were cultured alone or with breast cancer cells (mouse polyoma-middle T virus or primary human IDC) in 3D microfluidic platforms. Human monocytes and mouse macrophages in tumor cocultures exhibited significantly different FAD mean lifetimes and greater migration than monocultures at 24, 48, and 72 hours postseeding. In cocultures with primary human cancer cells, actively migrating monocyte-derived macrophages had greater redox ratios [NAD(P)H/FAD intensity] compared with passively migrating monocytes at 24 and 48 hours postseeding, reflecting metabolic heterogeneity in this subpopulation of monocytes. Genetic analyses further confirmed this metabolic heterogeneity. These results establish label-free autofluorescence imaging to quantify dynamic metabolism, polarization, and migration of macrophages at single-cell resolution within 3D microscale models. This combined culture and imaging system provides unique insights into spatiotemporal tumor-immune cross-talk within the 3D TME. SIGNIFICANCE: Label-free metabolic imaging and microscale culture technologies enable monitoring of single-cell macrophage metabolism, migration, and function in the 3D tumor microenvironment.
Collapse
Affiliation(s)
- Tiffany M Heaster
- Department of Biomedical Engineering, University of Wisconsin- Madison, Madison, Wisconsin.,Morgridge Institute for Research, Madison, Wisconsin
| | - Mouhita Humayun
- Department of Biomedical Engineering, University of Wisconsin- Madison, Madison, Wisconsin
| | - Jiaquan Yu
- Department of Biomedical Engineering, University of Wisconsin- Madison, Madison, Wisconsin.,Massachusetts Institute of Technology Koch Institute for Integrative Cancer Research, Cambridge, Massachusetts
| | - David J Beebe
- Department of Biomedical Engineering, University of Wisconsin- Madison, Madison, Wisconsin.,The University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, Wisconsin.,Department of Pathology & Laboratory Medicine, University of Wisconsin, Madison, Wisconsin
| | - Melissa C Skala
- Department of Biomedical Engineering, University of Wisconsin- Madison, Madison, Wisconsin. .,Morgridge Institute for Research, Madison, Wisconsin.,The University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, Wisconsin
| |
Collapse
|
31
|
Shang J, Wan F, Zhao L, Meng X, Li B. Potential Immunomodulatory Activity of a Selected Strain Bifidobacterium bifidum H3-R2 as Evidenced in vitro and in Immunosuppressed Mice. Front Microbiol 2020; 11:2089. [PMID: 32983062 PMCID: PMC7491056 DOI: 10.3389/fmicb.2020.02089] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 08/08/2020] [Indexed: 12/30/2022] Open
Abstract
The microbiota is directly involved in the development and modulation of the intestinal immune system. In particular, members of the genus Bifidobacterium play a primary role in immune regulation. In the present study, Bifidobacterium bifidum H3-R2 was screened from 15 bifidobacterium strains by in vitro experiment, showing a positive tolerance to digestive tract conditions, adhesion ability to intestinal epithelial cells and a regulatory effect on immune cell activity. Immunostimulatory activity of B. bifidum H3-R2 was also elucidated in vivo in cytoxan (CTX)-treated mice. The results showed that the administration of B. bifidum H3-R2 ameliorated the CTX-induced bodyweight loss and imbalanced expression of inflammatory cytokines, enhanced the production of secretory immunoglobulin A (SIgA), and promoted splenic lymphocyte proliferation, natural killer (NK) cell activity and phagocytosis of macrophages in immunosuppressed mice. In addition, B. bifidum H3-R2 restored injured intestinal mucosal, and increased the villus length and crypt depth in CTX-treated mice. The results could be helpful for understanding the functions of B. bifidum H3-R2, supporting its potential as a novel probiotic for immunoregulation.
Collapse
Affiliation(s)
- Jiacui Shang
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin, China
| | - Feng Wan
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin, China
| | - Le Zhao
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin, China
| | - Xiangchen Meng
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin, China.,School of Food Science, Northeast Agricultural University, Harbin, China
| | - Bailiang Li
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin, China.,School of Food Science, Northeast Agricultural University, Harbin, China
| |
Collapse
|
32
|
Xu F, Guo M, Huang W, Feng L, Zhu J, Luo K, Gao J, Zheng B, Kong LD, Pang T, Wu X, Xu Q. Annexin A5 regulates hepatic macrophage polarization via directly targeting PKM2 and ameliorates NASH. Redox Biol 2020; 36:101634. [PMID: 32863213 PMCID: PMC7369618 DOI: 10.1016/j.redox.2020.101634] [Citation(s) in RCA: 107] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 06/25/2020] [Accepted: 07/03/2020] [Indexed: 12/18/2022] Open
Abstract
Nonalcoholic steatohepatitis (NASH), the progressive form of nonalcoholic fatty liver disease (NAFLD), is becoming a common chronic liver disease with the characteristics of steatosis, inflammation and fibrosis. Macrophage plays an important role in the development of NASH. In this study, Annexin A5 (Anx A5) is identified with the special effect on hepatic macrophage phenotype shift from M1 to M2. And it is further demonstrated that Anx A5 significantly switches metabolic reprogramming from glycolysis to oxidative phosphorylation in activated macrophages. Mechanistically, the main target of Anx A5 in energy metabolism is confirmed to be pyruvate kinase M2 (PKM2). And we following reveal that Anx A5 directly interacts with PKM2 at ASP101, LEU104 and ARG106, inhibits phosphorylation of Y105, and promotes PKM2 tetramer formation. In addition, based on the results of PKM2 inhibitor (compound 3k) and the phosphorylated mutation (PKM2 (Y105E)), it is proved that Anx A5 exhibits the function in macrophage polarization dependently on PKM2 activity. In vivo studies also show that Anx A5 improves steatosis, inflammation and fibrosis in NASH mice due to specially regulating hepatic macrophages via interaction with PKM2. Therefore, we have revealed a novel function of Anx A5 in hepatic macrophage polarization and HFD-induced NASH, providing important insights into the metabolic reprogramming, which is important for NASH therapy.
Collapse
Affiliation(s)
- Fang Xu
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing Drum Tower Hospital, School of Life Sciences, Nanjing University, 163 Xianlin Avenue, Nanjing, 210023, China
| | - Mengmeng Guo
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing Drum Tower Hospital, School of Life Sciences, Nanjing University, 163 Xianlin Avenue, Nanjing, 210023, China
| | - Wei Huang
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing Drum Tower Hospital, School of Life Sciences, Nanjing University, 163 Xianlin Avenue, Nanjing, 210023, China
| | - Lili Feng
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing Drum Tower Hospital, School of Life Sciences, Nanjing University, 163 Xianlin Avenue, Nanjing, 210023, China
| | - Jiazhen Zhu
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing Drum Tower Hospital, School of Life Sciences, Nanjing University, 163 Xianlin Avenue, Nanjing, 210023, China
| | - Kangkang Luo
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing Drum Tower Hospital, School of Life Sciences, Nanjing University, 163 Xianlin Avenue, Nanjing, 210023, China
| | - Jian Gao
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing Drum Tower Hospital, School of Life Sciences, Nanjing University, 163 Xianlin Avenue, Nanjing, 210023, China
| | - Bingfeng Zheng
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing Drum Tower Hospital, School of Life Sciences, Nanjing University, 163 Xianlin Avenue, Nanjing, 210023, China
| | - Ling-Dong Kong
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing Drum Tower Hospital, School of Life Sciences, Nanjing University, 163 Xianlin Avenue, Nanjing, 210023, China
| | - Tao Pang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China
| | - Xudong Wu
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing Drum Tower Hospital, School of Life Sciences, Nanjing University, 163 Xianlin Avenue, Nanjing, 210023, China; State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China.
| | - Qiang Xu
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing Drum Tower Hospital, School of Life Sciences, Nanjing University, 163 Xianlin Avenue, Nanjing, 210023, China.
| |
Collapse
|
33
|
Varga Z, Molnár T, Mázló A, Kovács R, Jenei V, Kerekes K, Bácsi A, Koncz G. Differences in the sensitivity of classically and alternatively activated macrophages to TAK1 inhibitor-induced necroptosis. Cancer Immunol Immunother 2020; 69:2193-2207. [PMID: 32472370 PMCID: PMC7568718 DOI: 10.1007/s00262-020-02623-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 04/17/2020] [Indexed: 02/07/2023]
Abstract
Controlling the balance of pro-inflammatory M1 versus anti-inflammatory M2 macrophages may have paramount therapeutic benefit in cardiovascular diseases, infections, cancer and chronic inflammation. The targeted depletion of different macrophage populations provides a therapeutic option to regulate macrophage-mediated functions. Macrophages are highly sensitive to necroptosis, a newly described regulated cell death mediated by receptor-interacting serine/threonine-protein kinase 1 (RIPK1), RIPK3 and mixed lineage kinase domain like pseudokinase. Antagonists of inhibitors of apoptosis proteins (SMAC mimetics) block RIPK1 ubiquitination, while TGF-activated kinase 1 (TAK1) inhibitors prevent the phosphorylation of RIPK1, resulting in increased necroptosis. We compared the sensitivity of monocyte-derived human M1 and M2 cells to various apoptotic and necroptotic signals. The two cell types were equally sensitive to all investigated stimuli, but TAK1 inhibitor induced more intense necroptosis in M2 cells. Consequently, the treatment of co-cultured M1 and M2 cells with TAK1 inhibitor shifted the balance of the two populations toward M1 dominance. Blockage of either Aurora Kinase A or glycogen synthase kinase 3β, two newly described necroptosis inhibitors, increased the sensitivity of M1 cells to TAK1-inhibitor-induced cell death. Finally, we demonstrated that in vitro differentiated tumor-associated macrophages (TAM-like cells) were as highly sensitive to TAK1 inhibitor-induced necroptosis as M2 cells. Our results indicate that at least two different necroptotic pathways operate in macrophages and the targeted elimination of different macrophage populations by TAK1 inhibitor or SMAC mimetic may provide a therapeutic option to regulate the balance of inflammatory/anti-inflammatory macrophage functions.
Collapse
Affiliation(s)
- Zsófia Varga
- Department of Immunology, Faculty of Medicine, University of Debrecen, 1 Egyetem Square, Debrecen, 4032, Hungary.,Doctoral School of Molecular Cellular and Immune Biology, University of Debrecen, Debrecen, Hungary
| | - Tamás Molnár
- Department of Immunology, Faculty of Medicine, University of Debrecen, 1 Egyetem Square, Debrecen, 4032, Hungary.,Doctoral School of Molecular Cellular and Immune Biology, University of Debrecen, Debrecen, Hungary
| | - Anett Mázló
- Department of Immunology, Faculty of Medicine, University of Debrecen, 1 Egyetem Square, Debrecen, 4032, Hungary.,Doctoral School of Molecular Cellular and Immune Biology, University of Debrecen, Debrecen, Hungary.,MTA-DE Cell Biology and Signalling Research Group, University of Debrecen, Debrecen, Hungary
| | - Ramóna Kovács
- Department of Immunology, Faculty of Medicine, University of Debrecen, 1 Egyetem Square, Debrecen, 4032, Hungary.,Doctoral School of Molecular Cellular and Immune Biology, University of Debrecen, Debrecen, Hungary
| | - Viktória Jenei
- Department of Immunology, Faculty of Medicine, University of Debrecen, 1 Egyetem Square, Debrecen, 4032, Hungary
| | | | - Attila Bácsi
- Department of Immunology, Faculty of Medicine, University of Debrecen, 1 Egyetem Square, Debrecen, 4032, Hungary
| | - Gábor Koncz
- Department of Immunology, Faculty of Medicine, University of Debrecen, 1 Egyetem Square, Debrecen, 4032, Hungary.
| |
Collapse
|
34
|
RBM4 regulates M1 macrophages polarization through targeting STAT1-mediated glycolysis. Int Immunopharmacol 2020; 83:106432. [PMID: 32248017 DOI: 10.1016/j.intimp.2020.106432] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 03/11/2020] [Accepted: 03/19/2020] [Indexed: 12/20/2022]
Abstract
M1/M2 macrophages polarization play important roles in regulating tissue homeostasis. Recently, RNA-binding motif 4 (RBM4) has been reported to modulate the proliferation and expression of inflammatory factors in HeLa cells. However, whether RBM4 is involved in regulating macrophage polarization and inflammatory factor expression are still unknown. In this study, RAW264.7, a mouse macrophage cell line, were stimulated with interferon γ (IFN-γ) or interleukin-4 (IL-4) to induce M1/M2 macrophages polarization. We found that IFN-γ, but not IL-4, stimulation decreased RBM4 expression in macrophages, and RBM4 overexpression inhibits IFN-γ-induced M1 macrophage polarization. Furthermore, RNA-Sequencing, protein immunoprecipitation accompanied with mass spectrometry, and extracellular acidification rate analysis showed that RBM4 suppresses IFN-γ-induced M1 macrophage polarization though inhibiting glycolysis. Moreover, RBM4 knockdown promoted IFN-γ-induced signal transducer and activator of transcription 1 (STAT1) activation via increasing STAT1 mRNA stability, leading to the increase of glycolysis-related gene transcripts regulated by STAT1. Finally, we find that RBM4 interacts with YTH N6-methyladenosine RNA binding protein 2 (YTHDF2) to degrade m6A modified STAT1 mRNA, thereby regulating glycolysis and M1 macrophage polarization. Collectively, the current study firstly reports that RBM4 regulates M1 macrophages polarization through targeting STAT1-mediated glycolysis and shows that RBM4 is a possible candidate for regulating macrophage M1 polarization and inflammatory responses.
Collapse
|
35
|
Huang L, Nazarova EV, Russell DG. Mycobacterium tuberculosis. BACTERIA AND INTRACELLULARITY 2020:127-138. [DOI: 10.1128/9781683670261.ch9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Affiliation(s)
- Lu Huang
- Microbiology and Immunology, College of Veterinary Medicine; Cornell University; Ithaca NY 14853
| | - Evgeniya V. Nazarova
- Microbiology and Immunology, College of Veterinary Medicine; Cornell University; Ithaca NY 14853
| | - David G. Russell
- Microbiology and Immunology, College of Veterinary Medicine; Cornell University; Ithaca NY 14853
| |
Collapse
|
36
|
Zhao YY, Fu H, Liang XY, Zhang BL, Wei LL, Zhu JX, Chen MW, Zhao YF. Lipopolysaccharide inhibits GPR120 expression in macrophages via Toll-like receptor 4 and p38 MAPK activation. Cell Biol Int 2020; 44:89-97. [PMID: 31322778 DOI: 10.1002/cbin.11204] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 07/05/2019] [Indexed: 01/24/2023]
Abstract
Free fatty acid receptor G protein-coupled receptor 120 (GPR120) is highly expressed in macrophages and was reported to inhibit lipopolysaccharide (LPS)-stimulated cytokine expression. Under inflammation, macrophages exhibit striking functional changes, but changes in GPR120 expression and signaling are not known. In this study, the effects of LPS treatment on macrophage GPR120 expression and activation were investigated. The results showed that LPS inhibited GPR120 expression in mouse macrophage cell line Ana-1 cells. Moreover, LPS treatment inhibited GPR120 expression in mouse alveolar macrophages both in vitro and in vivo. The inhibitory effect of LPS on GPR120 expression was blocked by Toll-like receptor 4 (TLR4) inhibitor TAK242 and p38 mitogen-activated protein kinase inhibitor LY222820, but not by ERK1/2 inhibitor U0126 and c-Jun N-terminal kinase inhibitor SP600125. LPS-induced inhibition of GPR120 expression was not attenuated by GPR120 agonists TUG891 and GW9508. TUG891 inhibited the phagocytosis of alveolar macrophages, and LPS treatment counteracted the effects of TUG891 on phagocytosis. These results indicate that pretreatment with LPS inhibits GPR120 expression and activation in macrophages. It is suggested that LPS-induced inhibition of GPR120 expression is a reaction enhancing the LPS-induced pro-inflammatory response of macrophages.
Collapse
Affiliation(s)
- Yan-Yan Zhao
- Institute of Basic Medical Sciences, Xi'an Medical University, Xi'an, 710021, China
| | - Hui Fu
- Institute of Basic Medical Sciences, Xi'an Medical University, Xi'an, 710021, China
| | - Xiang-Yan Liang
- Institute of Basic Medical Sciences, Xi'an Medical University, Xi'an, 710021, China
| | - Bi-Lin Zhang
- Institute of Basic Medical Sciences, Xi'an Medical University, Xi'an, 710021, China
| | - Lan-Lan Wei
- Institute of Basic Medical Sciences, Xi'an Medical University, Xi'an, 710021, China
| | - Juan-Xia Zhu
- Institute of Basic Medical Sciences, Xi'an Medical University, Xi'an, 710021, China
| | - Ming-Wei Chen
- Shaanxi Provincial Research Center for Prevention and Treatment of Respiratory Diseases, Xi'an Medical University, Xi'an, 710021, China
| | - Yu-Feng Zhao
- Institute of Basic Medical Sciences, Xi'an Medical University, Xi'an, 710021, China
| |
Collapse
|
37
|
De Santa F, Vitiello L, Torcinaro A, Ferraro E. The Role of Metabolic Remodeling in Macrophage Polarization and Its Effect on Skeletal Muscle Regeneration. Antioxid Redox Signal 2019; 30:1553-1598. [PMID: 30070144 DOI: 10.1089/ars.2017.7420] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Significance: Macrophages are crucial for tissue homeostasis. Based on their activation, they might display classical/M1 or alternative/M2 phenotypes. M1 macrophages produce pro-inflammatory cytokines, reactive oxygen species (ROS), and nitric oxide (NO). M2 macrophages upregulate arginase-1 and reduce NO and ROS levels; they also release anti-inflammatory cytokines, growth factors, and polyamines, thus promoting angiogenesis and tissue healing. Moreover, M1 and M2 display key metabolic differences; M1 polarization is characterized by an enhancement in glycolysis and in the pentose phosphate pathway (PPP) along with a decreased oxidative phosphorylation (OxPhos), whereas M2 are characterized by an efficient OxPhos and reduced PPP. Recent Advances: The glutamine-related metabolism has been discovered as crucial for M2 polarization. Vice versa, flux discontinuities in the Krebs cycle are considered additional M1 features; they lead to increased levels of immunoresponsive gene 1 and itaconic acid, to isocitrate dehydrogenase 1-downregulation and to succinate, citrate, and isocitrate over-expression. Critical Issues: A macrophage classification problem, particularly in vivo, originating from a gap in the knowledge of the several intermediate polarization statuses between the M1 and M2 extremes, characterizes this field. Moreover, the detailed features of metabolic reprogramming crucial for macrophage polarization are largely unknown; in particular, the role of β-oxidation is highly controversial. Future Directions: Manipulating the metabolism to redirect macrophage polarization might be useful in various pathologies, including an efficient skeletal muscle regeneration. Unraveling the complexity pertaining to metabolic signatures that are specific for the different macrophage subsets is crucial for identifying new compounds that are able to trigger macrophage polarization and that might be used for therapeutical purposes.
Collapse
Affiliation(s)
- Francesca De Santa
- Institute of Cell Biology and Neurobiology (IBCN), National Research Council (CNR), Rome, Italy
| | - Laura Vitiello
- Laboratory of Pathophysiology of Cachexia and Metabolism of Skeletal Muscle, IRCCS San Raffaele Pisana, Rome, Italy
| | - Alessio Torcinaro
- Institute of Cell Biology and Neurobiology (IBCN), National Research Council (CNR), Rome, Italy.,Department of Biology and Biotechnology "Charles Darwin," Sapienza University, Rome, Italy
| | - Elisabetta Ferraro
- Laboratory of Pathophysiology of Cachexia and Metabolism of Skeletal Muscle, IRCCS San Raffaele Pisana, Rome, Italy
| |
Collapse
|
38
|
Fernandes DC, Eto SF, Moraes AC, Prado EJR, Medeiros ASR, Belo MAA, Samara SI, Costa PI, Pizauro JM. Phagolysosomal activity of macrophages in Nile tilapia (Oreochromis niloticus) infected in vitro by Aeromonas hydrophila: Infection and immunotherapy. FISH & SHELLFISH IMMUNOLOGY 2019; 87:51-61. [PMID: 30599256 DOI: 10.1016/j.fsi.2018.12.074] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 12/22/2018] [Accepted: 12/28/2018] [Indexed: 06/09/2023]
Abstract
The biochemical mechanisms involved in phagocytosis and the intracellular survival of Aeromonas hydrophila (Ah) in host macrophages (MΦs) are complex processes that affect infection success or failure. Thus, in the present study, we described the in vitro infection of Nile tilapia MΦs by a homologous bacterium and tested the effects of anti-A. hydrophila immunoglobulin Y (IgY) on the phagolysosomal activity and intracellular survival of the pathogen. The anti-Ah IgY modulated lysosomal acid phosphatase (LAP) activity as well as the production of reactive oxygen intermediates (ROIs) and nitric oxide (NO), thereby potentiating phagocytosis and the elimination of Ah. Thus, we assume that the specific IgY had a beneficial effect on infection control and postulated the use of the Nile tilapia MΦs as an important in vitro experimental model for the functional and therapeutic study of Ah infection.
Collapse
Affiliation(s)
- Dayanne C Fernandes
- Institute of Chemistry, Sao Paulo State University (Unesp), Araraquara, São Paulo, Brazil; Department of Technology, School of Agrarian and Veterinary Sciences, Sao Paulo State University (Unesp), Jaboticabal, Sao Paulo, Brazil
| | - Silas F Eto
- Department of Technology, School of Agrarian and Veterinary Sciences, Sao Paulo State University (Unesp), Jaboticabal, Sao Paulo, Brazil.
| | - Alessandra C Moraes
- Department of Preventive Veterinary Medicine of Unesp, School of Agrarian and Veterinary Sciences, Sao Paulo State University (Unesp), Jaboticabal, Sao Paulo, Brazil
| | - Ed Johnny R Prado
- Department of Preventive Veterinary Medicine of Unesp, School of Agrarian and Veterinary Sciences, Sao Paulo State University (Unesp), Jaboticabal, Sao Paulo, Brazil
| | - Andrea S R Medeiros
- Department of Preventive Veterinary Medicine of Unesp, School of Agrarian and Veterinary Sciences, Sao Paulo State University (Unesp), Jaboticabal, Sao Paulo, Brazil
| | - Marco A A Belo
- Department of Preventive Veterinary Medicine of Unesp, School of Agrarian and Veterinary Sciences, Sao Paulo State University (Unesp), Jaboticabal, Sao Paulo, Brazil
| | - Samir I Samara
- Department of Preventive Veterinary Medicine of Unesp, School of Agrarian and Veterinary Sciences, Sao Paulo State University (Unesp), Jaboticabal, Sao Paulo, Brazil
| | - Paulo I Costa
- Clinical Analysis Department, School of Pharmaceutical Sciences, São Paulo State University (Unesp), Araraquara, São Paulo, Brazil
| | - João M Pizauro
- Institute of Chemistry, Sao Paulo State University (Unesp), Araraquara, São Paulo, Brazil; Department of Technology, School of Agrarian and Veterinary Sciences, Sao Paulo State University (Unesp), Jaboticabal, Sao Paulo, Brazil
| |
Collapse
|
39
|
Huang L, Nazarova EV, Russell DG. Mycobacterium tuberculosis: Bacterial Fitness within the Host Macrophage. Microbiol Spectr 2019; 7:10.1128/microbiolspec.bai-0001-2019. [PMID: 30848232 PMCID: PMC6459685 DOI: 10.1128/microbiolspec.bai-0001-2019] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Indexed: 12/30/2022] Open
Abstract
Mycobacterium tuberculosis has evolved to become the single greatest cause of death from an infectious agent. The pathogen spends most of its infection cycle in its human host within a phagocyte. The bacterium has evolved to block the normal maturation and acidification of its phagosome and resides in a vacuole contiguous with the early endosomal network. Cytokine-mediated activation of the host cell can overcome this blockage, and an array of antimicrobial responses can limit its survival. The survival of M. tuberculosis in its host cell is fueled predominantly by fatty acids and cholesterol. The ability of M. tuberculosis to degrade sterols is an unusual metabolic characteristic that was likely retained from a saprophytic ancestor. Recent results with fluorescent M. tuberculosis reporter strains demonstrate that bacterial survival differs with the host macrophage population. Tissue-resident alveolar macrophages, which are biased towards an alternatively activated, M2-like phenotype, are more permissive to bacterial growth than monocyte-derived, inflammatory, M1-like interstitial macrophages. The differential growth of the bacterium in these different phagocyte populations appears to be linked to host cell metabolism.
Collapse
Affiliation(s)
- Lu Huang
- Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853
| | - Evgeniya V Nazarova
- Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853
| | - David G Russell
- Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853
| |
Collapse
|
40
|
Puchalska P, Martin SE, Huang X, Lengfeld JE, Daniel B, Graham MJ, Han X, Nagy L, Patti GJ, Crawford PA. Hepatocyte-Macrophage Acetoacetate Shuttle Protects against Tissue Fibrosis. Cell Metab 2019; 29:383-398.e7. [PMID: 30449686 PMCID: PMC6559243 DOI: 10.1016/j.cmet.2018.10.015] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 08/20/2018] [Accepted: 10/24/2018] [Indexed: 12/12/2022]
Abstract
Metabolic plasticity has been linked to polarized macrophage function, but mechanisms connecting specific fuels to tissue macrophage function remain unresolved. Here we apply a stable isotope tracing, mass spectrometry-based untargeted metabolomics approach to reveal the metabolome penetrated by hepatocyte-derived glucose and ketone bodies. In both classically and alternatively polarized macrophages, [13C]acetoacetate (AcAc) labeled ∼200 chemical features, but its reduced form D-[13C]β-hydroxybutyrate (D-βOHB) labeled almost none. [13C]glucose labeled ∼500 features, and while unlabeled AcAc competed with only ∼15% of them, the vast majority required the mitochondrial enzyme succinyl-coenzyme A-oxoacid transferase (SCOT). AcAc carbon labeled metabolites within the cytoplasmic glycosaminoglycan pathway, which regulates tissue fibrogenesis. Accordingly, livers of mice lacking SCOT in macrophages were predisposed to accelerated fibrogenesis. Exogenous AcAc, but not D-βOHB, ameliorated diet-induced hepatic fibrosis. These data support a hepatocyte-macrophage ketone shuttle that segregates AcAc from D-βOHB, coordinating the fibrogenic response to hepatic injury via mitochondrial metabolism in tissue macrophages.
Collapse
Affiliation(s)
- Patrycja Puchalska
- Division of Molecular Medicine, Department of Medicine, University of Minnesota, 401 East River Parkway, MMC 194, Minneapolis, MN 55455, USA; Center for Metabolic Origins of Disease, Sanford Burnham Prebys Medical Discovery Institute, Orlando, FL 32827, USA
| | - Shannon E Martin
- Center for Metabolic Origins of Disease, Sanford Burnham Prebys Medical Discovery Institute, Orlando, FL 32827, USA; Pathobiology Graduate Program, Brown University, Providence, RI 02912, USA
| | - Xiaojing Huang
- Center for Metabolic Origins of Disease, Sanford Burnham Prebys Medical Discovery Institute, Orlando, FL 32827, USA; Department of Chemistry, Washington University, St. Louis, MO 63110, USA; Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Justin E Lengfeld
- Division of Molecular Medicine, Department of Medicine, University of Minnesota, 401 East River Parkway, MMC 194, Minneapolis, MN 55455, USA
| | - Bence Daniel
- Center for Metabolic Origins of Disease, Sanford Burnham Prebys Medical Discovery Institute, Orlando, FL 32827, USA; Department of Medicine, Johns Hopkins University School of Medicine, Johns Hopkins All Children's Hospital, Saint Petersburg, FL 33701, USA
| | | | - Xianlin Han
- Center for Metabolic Origins of Disease, Sanford Burnham Prebys Medical Discovery Institute, Orlando, FL 32827, USA; Barshop Institute for Longevity and Aging Studies, Department of Medicine, Division of Diabetes, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Laszlo Nagy
- Center for Metabolic Origins of Disease, Sanford Burnham Prebys Medical Discovery Institute, Orlando, FL 32827, USA; Department of Medicine, Johns Hopkins University School of Medicine, Johns Hopkins All Children's Hospital, Saint Petersburg, FL 33701, USA; Department of Biological Chemistry, Johns Hopkins University School of Medicine, Johns Hopkins All Children's Hospital, Saint Petersburg, FL 33701, USA
| | - Gary J Patti
- Department of Chemistry, Washington University, St. Louis, MO 63110, USA
| | - Peter A Crawford
- Division of Molecular Medicine, Department of Medicine, University of Minnesota, 401 East River Parkway, MMC 194, Minneapolis, MN 55455, USA; Center for Metabolic Origins of Disease, Sanford Burnham Prebys Medical Discovery Institute, Orlando, FL 32827, USA; Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
41
|
Morón-Calvente V, Romero-Pinedo S, Toribio-Castelló S, Plaza-Díaz J, Abadía-Molina AC, Rojas-Barros DI, Beug ST, LaCasse EC, MacKenzie A, Korneluk R, Abadía-Molina F. Inhibitor of apoptosis proteins, NAIP, cIAP1 and cIAP2 expression during macrophage differentiation and M1/M2 polarization. PLoS One 2018. [PMID: 29518103 PMCID: PMC5843221 DOI: 10.1371/journal.pone.0193643] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Monocytes and macrophages constitute the first line of defense of the immune system against external pathogens. Macrophages have a highly plastic phenotype depending on environmental conditions; the extremes of this phenotypic spectrum are a pro-inflammatory defensive role (M1 phenotype) and an anti-inflammatory tissue-repair one (M2 phenotype). The Inhibitor of Apoptosis (IAP) proteins have important roles in the regulation of several cellular processes, including innate and adaptive immunity. In this study we have analyzed the differential expression of the IAPs, NAIP, cIAP1 and cIAP2, during macrophage differentiation and polarization into M1 or M2. In polarized THP-1 cells and primary human macrophages, NAIP is abundantly expressed in M2 macrophages, while cIAP1 and cIAP2 show an inverse pattern of expression in polarized macrophages, with elevated expression levels of cIAP1 in M2 and cIAP2 preferentially expressed in M1. Interestingly, treatment with the IAP antagonist SMC-LCL161, induced the upregulation of NAIP in M2, the downregulation of cIAP1 in M1 and M2 and an induction of cIAP2 in M1 macrophages.
Collapse
Affiliation(s)
- Virginia Morón-Calvente
- Department of Cell Biology, University of Granada, Granada, Spain
- Biomedical Research Centre, University of Granada, Granada, Spain
| | - Salvador Romero-Pinedo
- Biomedical Research Centre, University of Granada, Granada, Spain
- Department of Biochemistry and Molecular Biology III and Immunology, University of Granada, Granada, Spain
| | | | - Julio Plaza-Díaz
- Biomedical Research Centre, University of Granada, Granada, Spain
- Department of Biochemistry and Molecular Biology II, University of Granada, Granada, Spain
- Institute of Nutrition and Food Technology “José Mataix”, University of Granada, Granada, Spain
| | - Ana C. Abadía-Molina
- Biomedical Research Centre, University of Granada, Granada, Spain
- Department of Biochemistry and Molecular Biology III and Immunology, University of Granada, Granada, Spain
| | - Domingo I. Rojas-Barros
- Institute of Parasitology and Biomedicine “López-Neyra”, Spanish National Research Council (CSIC), Granada, Spain
| | - Shawn T. Beug
- Apoptosis Research Centre, Children’s Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa ON, Canada
| | - Eric C. LaCasse
- Apoptosis Research Centre, Children’s Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa ON, Canada
| | - Alex MacKenzie
- Apoptosis Research Centre, Children’s Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa ON, Canada
- Department of Pediatrics, University of Ottawa, Ottawa ON, Canada
| | - Robert Korneluk
- Apoptosis Research Centre, Children’s Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa ON, Canada
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa ON, Canada
| | - Francisco Abadía-Molina
- Department of Cell Biology, University of Granada, Granada, Spain
- Biomedical Research Centre, University of Granada, Granada, Spain
- Institute of Nutrition and Food Technology “José Mataix”, University of Granada, Granada, Spain
- * E-mail:
| |
Collapse
|
42
|
Wang Y, Wu T, Hu D, Weng X, Wang X, Chen PJ, Luo X, Wang H, Ning Q. Intracellular hepatitis B virus increases hepatic cholesterol deposition in alcoholic fatty liver via hepatitis B core protein. J Lipid Res 2017; 59:58-68. [PMID: 29133292 PMCID: PMC5748497 DOI: 10.1194/jlr.m079533] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 10/23/2017] [Indexed: 12/13/2022] Open
Abstract
Hepatitis B virus (HBV) infection is a prevalent infectious disease with serious outcomes like chronic and acute hepatitis, cirrhosis, and hepatocellular carcinoma. However, the metabolic alteration by HBV is rarely taken into consideration. With the high prevalence of alcohol consumption and chronic HBV infection, their overlap is assumed to be an increasing latent hazard; although the extent has not been calculated. Moreover, the impact of chronic alcohol consumption combined with HBV on cholesterol metabolism is unknown. Six-week-old male FVB/Ncrl mice were hydrodynamically injected with a pGEM-4Z-1.3HBV vector and then fed an ethanol diet for 6 weeks. Serum biomarkers and liver histology, liver cholesterol levels, and cholesterol metabolism-related molecules were measured. In vitro assays with HBx, hepatitis B surface (HBs), or hepatitis B core (HBc) protein expression in HepG2 cells costimulated with ethanol were conducted to assess the cholesterol metabolism. HBV expression synergistically increased cholesterol deposition in the setting of alcoholic fatty liver. The increase of intrahepatic cholesterol was due to metabolic alteration in cholesterol metabolism, including increased cholesterol synthesis, decreased cholesterol degradation, and impaired cholesterol uptake. Overexpression of HBV component HBc, but not HBs or HBx, selectively promoted the hepatocellular cholesterol level.
Collapse
Affiliation(s)
- Yaqi Wang
- Department and Institute of Infectious Disease Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ting Wu
- Department and Institute of Infectious Disease Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Danqing Hu
- Department and Institute of Infectious Disease Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xinxin Weng
- Department and Institute of Infectious Disease Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiaojing Wang
- Department and Institute of Infectious Disease Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Pei-Jer Chen
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Xiaoping Luo
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Hongwu Wang
- Department and Institute of Infectious Disease Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qin Ning
- Department and Institute of Infectious Disease Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
43
|
Beneke A, Guentsch A, Hillemann A, Zieseniss A, Swain L, Katschinski DM. Loss of PHD3 in myeloid cells dampens the inflammatory response and fibrosis after hind-limb ischemia. Cell Death Dis 2017; 8:e2976. [PMID: 28796258 PMCID: PMC5596563 DOI: 10.1038/cddis.2017.375] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 06/22/2017] [Accepted: 07/04/2017] [Indexed: 02/06/2023]
Abstract
Macrophages are essential for the inflammatory response after an ischemic insult and thereby influence tissue recovery. For the oxygen sensing prolyl-4-hydroxylase domain enzyme (PHD) 2 a clear impact on the macrophage-mediated arteriogenic response after hind-limb ischemia has been demonstrated previously, which involves fine tuning a M2-like macrophage population. To analyze the role of PHD3 in macrophages, we performed hind-limb ischemia (ligation and excision of the femoral artery) in myeloid-specific PHD3 knockout mice (PHD3−/−) and analyzed the inflammatory cell invasion, reperfusion recovery and fibrosis in the ischemic muscle post-surgery. In contrast to PHD2, reperfusion recovery and angiogenesis was unaltered in PHD3−/− compared to WT mice. Macrophages from PHD3−/− mice showed, however, a dampened inflammatory reaction in the affected skeletal muscle tissues compared to WT controls. This was associated with a decrease in fibrosis and an anti-inflammatory phenotype of the PHD3−/− macrophages, as well as decreased expression of Cyp2s1 and increased PGE2-secretion, which could be mimicked by PHD3−/− bone marrow-derived macrophages in serum starvation.
Collapse
Affiliation(s)
- Angelika Beneke
- Institute of Cardiovascular Physiology, University Medical Center, Göttingen, Germany
| | - Annemarie Guentsch
- Institute of Cardiovascular Physiology, University Medical Center, Göttingen, Germany
| | - Annette Hillemann
- Institute of Cardiovascular Physiology, University Medical Center, Göttingen, Germany
| | - Anke Zieseniss
- Institute of Cardiovascular Physiology, University Medical Center, Göttingen, Germany
| | - Lija Swain
- Institute of Cardiovascular Physiology, University Medical Center, Göttingen, Germany
| | - Dörthe M Katschinski
- Institute of Cardiovascular Physiology, University Medical Center, Göttingen, Germany
| |
Collapse
|
44
|
Li M, Qian M, Xu J. Vascular Endothelial Regulation of Obesity-Associated Insulin Resistance. Front Cardiovasc Med 2017; 4:51. [PMID: 28848738 PMCID: PMC5552760 DOI: 10.3389/fcvm.2017.00051] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 07/27/2017] [Indexed: 12/24/2022] Open
Abstract
Obesity is a worldwide epidemic that predisposes individuals to metabolic complications, such as type 2 diabetes mellitus and non-alcoholic fatty liver disease, all of which are related to an imbalance between food intake and energy expenditure. Identification of the pathogenic molecular mechanisms and effective therapeutic approaches are urgently needed. A well-accepted paradigm is that crosstalk between organs/tissues contributes to diseases. Endothelial dysfunction characterizes metabolic disorders and the related vascular complications. Over the past two decades, overwhelming studies have focused on mechanisms that lead to endothelial dysfunction. New investigations, however, have begun to appreciate the opposite direction of the crosstalk: endothelial regulation of metabolism, although the underlying mechanisms remain to be elucidated. This review summarizes the evidence that supports the concept of endothelial regulation of obesity and the associated insulin resistance in fat, liver, and skeletal muscles, the classic targets of insulin. Outstanding questions and future research directions are highlighted. Identification of the mechanisms of vascular endothelial regulation of metabolism may offer strategies for prevention and treatment of obesity and the related metabolic complications.
Collapse
Affiliation(s)
- Manna Li
- Department of Medicine, Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Ming Qian
- Department of Medicine, Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Jian Xu
- Department of Medicine, Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| |
Collapse
|