1
|
Li Y, Xu X, Wu X, Li J, Chen S, Chen D, Li G, Tang Z. Cell polarization in ischemic stroke: molecular mechanisms and advances. Neural Regen Res 2025; 20:632-645. [PMID: 38886930 PMCID: PMC11433909 DOI: 10.4103/nrr.nrr-d-23-01336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 10/23/2023] [Accepted: 12/18/2023] [Indexed: 06/20/2024] Open
Abstract
Ischemic stroke is a cerebrovascular disease associated with high mortality and disability rates. Since the inflammation and immune response play a central role in driving ischemic damage, it becomes essential to modulate excessive inflammatory reactions to promote cell survival and facilitate tissue repair around the injury site. Various cell types are involved in the inflammatory response, including microglia, astrocytes, and neutrophils, each exhibiting distinct phenotypic profiles upon stimulation. They display either proinflammatory or anti-inflammatory states, a phenomenon known as 'cell polarization.' There are two cell polarization therapy strategies. The first involves inducing cells into a neuroprotective phenotype in vitro, then reintroducing them autologously. The second approach utilizes small molecular substances to directly affect cells in vivo. In this review, we elucidate the polarization dynamics of the three reactive cell populations (microglia, astrocytes, and neutrophils) in the context of ischemic stroke, and provide a comprehensive summary of the molecular mechanisms involved in their phenotypic switching. By unraveling the complexity of cell polarization, we hope to offer insights for future research on neuroinflammation and novel therapeutic strategies for ischemic stroke.
Collapse
Affiliation(s)
- Yuanwei Li
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Xiaoxiao Xu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Xuan Wu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Jiarui Li
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Shiling Chen
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Danyang Chen
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Gaigai Li
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Zhouping Tang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| |
Collapse
|
2
|
Lewis CV, Garcia AM, Burciaga SD, Posey JN, Jordan M, Nguyen TTN, Stenmark KR, Mickael C, Sul C, Delaney C, Nozik ES. Redistribution of SOD3 expression due to R213G polymorphism affects pulmonary interstitial macrophage reprogramming in response to hypoxia. Physiol Genomics 2024; 56:776-790. [PMID: 39311838 DOI: 10.1152/physiolgenomics.00078.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 08/14/2024] [Accepted: 09/11/2024] [Indexed: 10/25/2024] Open
Abstract
The extracellular isoform of superoxide dismutase (SOD3) is decreased in patients and animals with pulmonary hypertension (PH). The human R213G single-nucleotide polymorphism (SNP) in SOD3 causes its release from tissue extracellular matrix (ECM) into extracellular fluids, without modulating enzyme activity, increasing cardiovascular disease risk in humans and exacerbating chronic hypoxic PH in mice. Given the importance of interstitial macrophages (IMs) to PH pathogenesis, this study aimed to determine whether R213G SOD3 increases IM accumulation and alters IM reprogramming in response to hypoxia. R213G mice and wild-type (WT) controls were exposed to hypobaric hypoxia for 4 or 14 days compared with normoxia. Flow cytometry demonstrated a transient increase in IMs at day 4 in both strains. Contrary to our hypothesis, the R213G SNP did not augment IM accumulation. To determine strain differences in the IM reprogramming response to hypoxia, we performed RNAsequencing on IMs isolated at each timepoint. We found that IMs from R213G mice exposed to hypoxia activated ECM-related pathways and a combination of alternative macrophage and proinflammatory signaling. Furthermore, when compared with WT responses, IMs from R213G mice lacked metabolic remodeling and demonstrated a blunted anti-inflammatory response between the early (day 4) and later (day 14) timepoints. We confirmed metabolic responses using Agilent Seahorse assays, whereby WT, but not R213G, IMs upregulated glycolysis at day 4 that returned to baseline at day 14. Finally, we identify differential regulation of several redox-sensitive upstream regulators that could be investigated in future studies.NEW & NOTEWORTHY Redistributed expression of SOD3 out of tissue ECM due to the human R213G SNP exacerbates chronic hypoxic PH. Highlighting the importance of macrophage phenotype, our findings reveal that the R213G SNP does not exacerbate pulmonary macrophage accumulation in response to hypoxia but influences their metabolic and phenotypic reprogramming. We demonstrate a deficiency in the metabolic response to hypoxic stress in R213G macrophages, associated with weakened inflammatory resolution and activation of profibrotic pathways implicated in PH.
Collapse
Affiliation(s)
- Caitlin V Lewis
- Cardiovascular Pulmonary Research Laboratories, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Section of Pediatric Critical Care Medicine, Department of Pediatrics, Children's Hospital Colorado, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Anastacia M Garcia
- Division of Cardiology, Department of Pediatrics, Children's Hospital Colorado, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Samuel D Burciaga
- Cardiovascular Pulmonary Research Laboratories, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Section of Pediatric Critical Care Medicine, Department of Pediatrics, Children's Hospital Colorado, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Janelle N Posey
- Cardiovascular Pulmonary Research Laboratories, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Section of Neonatology, Department of Pediatrics, Division of Cardiology, Children's Hospital Colorado, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Mariah Jordan
- Cardiovascular Pulmonary Research Laboratories, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Section of Neonatology, Department of Pediatrics, Division of Cardiology, Children's Hospital Colorado, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Thi-Tina N Nguyen
- Cardiovascular Pulmonary Research Laboratories, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Section of Pediatric Critical Care Medicine, Department of Pediatrics, Children's Hospital Colorado, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Kurt R Stenmark
- Cardiovascular Pulmonary Research Laboratories, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Section of Pediatric Critical Care Medicine, Department of Pediatrics, Children's Hospital Colorado, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Claudia Mickael
- Cardiovascular Pulmonary Research Laboratories, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Section of Pediatric Critical Care Medicine, Department of Pediatrics, Children's Hospital Colorado, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Christina Sul
- Cardiovascular Pulmonary Research Laboratories, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Section of Pediatric Critical Care Medicine, Department of Pediatrics, Children's Hospital Colorado, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Cassidy Delaney
- Cardiovascular Pulmonary Research Laboratories, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Section of Neonatology, Department of Pediatrics, Division of Cardiology, Children's Hospital Colorado, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Eva S Nozik
- Cardiovascular Pulmonary Research Laboratories, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Section of Pediatric Critical Care Medicine, Department of Pediatrics, Children's Hospital Colorado, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| |
Collapse
|
3
|
Lin ZH, Lo HC, Chang CC, Lu MK, Tseng AJ, Chao CH, Chao CH, Lin TY. Sulfated polysaccharide from Antrodia cinnamomea mycelium cultured with zinc sulfate stimulates M1 polarization of macrophages through AKT/mTOR pathways. Int J Biol Macromol 2024; 279:135548. [PMID: 39270905 DOI: 10.1016/j.ijbiomac.2024.135548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 08/30/2024] [Accepted: 09/09/2024] [Indexed: 09/15/2024]
Abstract
Antrodia cinnamomea-derived sulfated polysaccharides (Ac-SPSs) have health benefits, but their yield is low. This study explores a strategy to increase Ac-SPS yield and elucidates the biofunctions of Ac-SPS. For this, A. cinnamomea mycelia were treated with zinc sulfate (ZnSO4) administered at 1, 10, and 100 μM. Firstly, functional assay indicated that ZnSO4 increases the Ac-SPS yield by 20 %-30 % compared with the control treatment. ZnSO4 engenders a population of middle-molecular-weight (~200 kDa) Ac-SPSs. Ac-SPS (ASZ-10) from A. cinnamomea treated with 10 μM ZnSO4 exhibits the best anti-proliferation ability against lung cancer A549 cells. Co-treatment of ASZ-10 does not inhibit lipopolysaccharide-induced inflammation but does induce M1-related markers of macrophage RAW264.7 cells. Secondly, immunomodulatory properties showed that ASZ-10 increases the expression of CD80+ and CD86+ in M-CSF-stimulated bone-marrow-derived macrophages. ASZ-10 induces M1 polarization through up-regulation of the AKT/mTOR pathway as confirmed by AKT and mTOR inhibitors eliminating ASZ-10-induced M1-like markers of macrophages. Through systemic chemical and functional analysis, this study shows that trace amounts (10 μM) of ZnSO4 increase Ac-SPS yield and it reveals that ASZ-10 exhibits anti-cancer activity and acts as a stimulator for M1 macrophages by stimulation of AKT and mTOR.
Collapse
Affiliation(s)
- Zhi-Hu Lin
- Institute of Traditional Medicine, National Yang Ming Chiao Tung University, 155 Li-Nung St., Sec. 2, Shipai, Beitou, Taipei 112, Taiwan
| | - Hung-Chih Lo
- Institute of Traditional Medicine, National Yang Ming Chiao Tung University, 155 Li-Nung St., Sec. 2, Shipai, Beitou, Taipei 112, Taiwan; Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei, Taiwan
| | - Chia-Chuan Chang
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Mei-Kuang Lu
- Institute of Traditional Medicine, National Yang Ming Chiao Tung University, 155 Li-Nung St., Sec. 2, Shipai, Beitou, Taipei 112, Taiwan; School of Chinese Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; National Research Institute of Chinese Medicine, Ministry of Health and Welfare, 155-1 Li-Nung St., Sec. 2, Shipai, Beitou, Taipei 112, Taiwan; Traditional Chinese Medicine Glycomics Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| | - Ai-Jung Tseng
- Institute of Traditional Medicine, National Yang Ming Chiao Tung University, 155 Li-Nung St., Sec. 2, Shipai, Beitou, Taipei 112, Taiwan
| | - Chi-Hsein Chao
- School of Chinese Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chi-Hong Chao
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu, Taiwan; Center for Intelligent Drug Systems and Smart Bio-devices (IDS2B), National Yang Ming Chiao Tung University, Hsinchu, Taiwan
| | - Tung-Yi Lin
- Institute of Traditional Medicine, National Yang Ming Chiao Tung University, 155 Li-Nung St., Sec. 2, Shipai, Beitou, Taipei 112, Taiwan; Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei, Taiwan; School of Chinese Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Traditional Chinese Medicine Glycomics Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| |
Collapse
|
4
|
Yan J, Zhang X, Wang H, Jia X, Wang R, Wu S, Zhu ZJ, Tan M, Horng T. Macrophage NRF1 promotes mitochondrial protein turnover via the ubiquitin proteasome system to limit mitochondrial stress and inflammation. Cell Rep 2024; 43:114780. [PMID: 39325625 DOI: 10.1016/j.celrep.2024.114780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 08/09/2024] [Accepted: 09/05/2024] [Indexed: 09/28/2024] Open
Abstract
Macrophage elaboration of inflammatory responses is dynamically regulated, shifting from acute induction to delayed suppression during the course of infection. Here, we show that such regulation of inflammation is modulated by dynamic shifts in metabolism. In macrophages exposed to the bacterial product lipopolysaccharide (LPS), an initial induction of protein biosynthesis is followed by compensatory induction of the transcription factor nuclear factor erythroid 2-like 1 (NRF1), leading to increased flux through the ubiquitin proteasome system (UPS). A major target of NRF1-mediated UPS flux is the mitochondrial proteome, and in the absence of NRF1, ubiquitinated mitochondrial proteins accumulate to trigger severe mitochondrial stress. Such mitochondrial stress engages the integrated stress response-ATF4 axis, which limits mitochondrial translation to attenuate mitochondrial stress but amplifies inflammatory responses to augment susceptibility to septic shock. Therefore, NRF1 mediates a dynamic regulation of mitochondrial proteostasis in inflammatory macrophages that contributes to curbing inflammatory responses.
Collapse
Affiliation(s)
- Jiawei Yan
- School of Life Sciences and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Xin Zhang
- School of Life Sciences and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Huiying Wang
- School of Life Sciences and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Xinglong Jia
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Ruohong Wang
- University of Chinese Academy of Sciences, Beijing 100049, China; Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 200032, China
| | - Shuangyang Wu
- Gregor Mendel Institute of Molecular Plant Biology, Austrian Academy of Sciences, Dr. Bohr-Gasse 3, 1030 Vienna, Austria
| | - Zheng-Jiang Zhu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 200032, China; Shanghai Key Laboratory of Aging Studies, Shanghai 201210, China
| | - Minjia Tan
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Tiffany Horng
- School of Life Sciences and Technology, ShanghaiTech University, Shanghai 201210, China.
| |
Collapse
|
5
|
Du Y, Zhang Y, Jiang Z, Xu L, Ru J, Wei S, Chen W, Dong R, Zhang S, Jia T. Triptolide alleviates acute gouty arthritis caused by monosodium urate crystals by modulating macrophage polarization and neutrophil activity. Immunol Lett 2024; 269:106907. [PMID: 39122094 DOI: 10.1016/j.imlet.2024.106907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 07/16/2024] [Accepted: 08/07/2024] [Indexed: 08/12/2024]
Abstract
The present study focused on the efficacy and role of triptolide (TPL) in relieving symptoms of acute gouty arthritis (AGA) in vivo and in vitro. The effects of TPL in AGA were investigated in monosodium urate (MSU)-treated rat ankles, RAW264.7 macrophages, and neutrophils isolated from mouse peritoneal cavity. Observation of pathological changes in the ankle joint of rats. Enzyme-linked immunosorbent assay and real-time quantitative polymerase chain reaction (RT-qPCR) were performed to detect the expression levels of inflammatory factors and chemokines. The levels of the indicators of macrophage M1/M2 polarization, and the mechanistic targets of Akt and rapamycin complex 2, were determined via western blotting and RT-qPCR. The expression levels of CD86 and CD206 were detected using immunohistochemistry. Neutrophil migration was observed via air pouch experiments in vivo and Transwell cell migration assay in vitro. Myeloperoxidase (MPO) and Neutrophil elastase (NE) release was analyzed by via immunohistochemistry and immunofluorescence. The expression levels of beclin-1, LC3B, Bax, Bcl-2, and cleaved caspase-3 in neutrophils were determined via western blotting and immunofluorescence. Neutrophil apoptosis was detected using the terminal deoxynucleotidyl transferase dUTP nick end labeling assay. Our results suggest that TPL inhibited inflammatory cell infiltration in rat ankle joints and inflammatory factor and chemokine secretion in rat serum, regulated macrophage polarization through the PI3K/AKT signaling pathway, suppressed inflammatory factor and chemokine expression in neutrophils, and inhibited neutrophil migration, neutrophil extracellular trap formation, transitional autophagy, and apoptosis. This suggests that TPL can prevent and treat MSU-induced AGA by regulating macrophage polarization through the PI3K/Akt pathway and modulating neutrophil activity.
Collapse
Affiliation(s)
- Yan Du
- Faculty of Basic Medicine, Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan 650500, China
| | - Yurong Zhang
- Faculty of Basic Medicine, Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan 650500, China
| | - Zhuxin Jiang
- Faculty of Basic Medicine, Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan 650500, China
| | - Lianjie Xu
- Faculty of Basic Medicine, Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan 650500, China
| | - Jing Ru
- Faculty of Basic Medicine, Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan 650500, China
| | - Shanshan Wei
- Faculty of Basic Medicine, Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan 650500, China
| | - Wenhui Chen
- Faculty of Basic Medicine, Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan 650500, China; Yunnan Provincial Key Laboratory of Molecular Biology for Sinomedicine, Kunming, Yunnan 450500, , China; Key Laboratory of Microcosmic Syndrome Differentiation, Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan 450500, , China
| | - Renjie Dong
- Faculty of Basic Medicine, Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan 650500, China
| | - Shan Zhang
- Faculty of Basic Medicine, Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan 650500, China; Yunnan Provincial Key Laboratory of Molecular Biology for Sinomedicine, Kunming, Yunnan 450500, , China; Key Laboratory of Microcosmic Syndrome Differentiation, Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan 450500, , China.
| | - Tao Jia
- Department of Orthopedics, First Clinical Medical College of Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan 650021, , China.
| |
Collapse
|
6
|
Wang J, Wu Q, Wang X, Liu H, Chen M, Xu L, Zhang Z, Li K, Li W, Zhong J. Targeting Macrophage Phenotypes and Metabolism as Novel Therapeutic Approaches in Atherosclerosis and Related Cardiovascular Diseases. Curr Atheroscler Rep 2024; 26:573-588. [PMID: 39133247 PMCID: PMC11392985 DOI: 10.1007/s11883-024-01229-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/22/2024] [Indexed: 08/13/2024]
Abstract
PURPOSE OF THE REVIEW Macrophage accumulation and activation function as hallmarks of atherosclerosis and have complex and intricate dynamics throughout all components and stages of atherosclerotic plaques. In this review, we focus on the regulatory roles and underlying mechanisms of macrophage phenotypes and metabolism in atherosclerosis. We highlight the diverse range of macrophage phenotypes present in atherosclerosis and their potential roles in progression and regression of atherosclerotic plaque. Furthermore, we discuss the challenges and opportunities in developing therapeutic strategies for preventing and treating atherosclerotic cardiovascular disease. RECENT FINDINGS Dysregulation of macrophage polarization between the proinflammatory M1 and anti-inflammatory M2 phenotypealters the immuno-inflammatory response during atherosclerosis progression, leading to plaque initiation, growth, and ultimately rupture. Altered metabolism of macrophage is a key feature for their function and the subsequent progression of atherosclerotic cardiovascular disease. The immunometabolism of macrophage has been implicated to macrophage activation and metabolic rewiring of macrophages within atherosclerotic lesions, thereby shifting altered macrophage immune-effector and tissue-reparative function. Targeting macrophage phenotypes and metabolism are potential therapeutic strategies in the prevention and treatment of atherosclerosis and atherosclerotic cardiovascular diseases. Understanding the precise function and metabolism of specific macrophage subsets and their contributions to the composition and growth of atherosclerotic plaques could reveal novel strategies to delay or halt development of atherosclerotic cardiovascular diseases and their associated pathophysiological consequences. Identifying biological stimuli capable of modulating macrophage phenotypes and metabolism may lead to the development of innovative therapeutic approaches for treating patients with atherosclerosis and coronary artery diseases.
Collapse
Affiliation(s)
- Juan Wang
- Beijing Key Laboratory of Hypertension, Heart Center of Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China.
| | - Qiang Wu
- Senior Department of Cardiology, the Sixth Medical Center, Chinese PLA General Hospital, Beijing, China
- Journal of Geriatric Cardiology Editorial Office, Chinese PLA General Hospital, Beijing, China
| | - Xinyu Wang
- Beijing Key Laboratory of Hypertension, Heart Center of Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Hongbin Liu
- Department of Cardiology, the Second Medical Center, National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China
| | - Mulei Chen
- Beijing Key Laboratory of Hypertension, Heart Center of Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Li Xu
- Beijing Key Laboratory of Hypertension, Heart Center of Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Ze Zhang
- National Institute of Biological Sciences, Beijing, China
| | - Kuibao Li
- Beijing Key Laboratory of Hypertension, Heart Center of Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Weiming Li
- Beijing Key Laboratory of Hypertension, Heart Center of Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China.
| | - Jiuchang Zhong
- Beijing Key Laboratory of Hypertension, Heart Center of Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
7
|
Ma H, Gao L, Chang R, Zhai L, Zhao Y. Crosstalk between macrophages and immunometabolism and their potential roles in tissue repair and regeneration. Heliyon 2024; 10:e38018. [PMID: 39381218 PMCID: PMC11458987 DOI: 10.1016/j.heliyon.2024.e38018] [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: 05/31/2024] [Revised: 09/16/2024] [Accepted: 09/16/2024] [Indexed: 10/10/2024] Open
Abstract
Immune metabolism is a result of many specific metabolic reactions, such as glycolysis, the tricarboxylic acid (TCA) pathway, the pentose phosphate pathway (PPP), mitochondrial oxidative phosphorylation (OXPHOS), fatty acid oxidation (FAO), fatty acid biosynthesis (FAs) and amino acid pathways, which promote cell proliferation and maintenance with structural and pathological energy to regulate cellular signaling. The metabolism of macrophages produces many metabolic intermediates that play important regulatory roles in tissue repair and regeneration. The metabolic activity of proinflammatory macrophages (M1) mainly depends on glycolysis and the TCA cycle system, but anti-inflammatory macrophages (M2) have intact functions of the TCA cycle, which enhances FAO and is dependent on OXPHOS. However, the metabolic mechanisms of macrophages in tissue repair and regeneration have not been well investigated. Thus, we review how three main metabolic mechanisms of macrophages, glucose metabolism, lipid metabolism, and amino acid metabolism, regulate tissue repair and regeneration.
Collapse
Affiliation(s)
- Hongbo Ma
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 610075, China
| | - Limei Gao
- Department of Cardiovascular Medicine, Shenzhen Longhua District Central Hospital, Shenzhen, 518110, China
| | - Rong Chang
- Department of Cardiovascular Medicine, Shenzhen Longhua District Central Hospital, Shenzhen, 518110, China
| | - Lihong Zhai
- Institute of Neuroscience and Brain Disease, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, 441000, Hubei, China
| | - Yanli Zhao
- Department of Cardiovascular Medicine, Shenzhen Longhua District Central Hospital, Shenzhen, 518110, China
- Department of Medical Laboratory, Shenzhen Longhua District Central Hospital, Shenzhen, 518110, China
| |
Collapse
|
8
|
Zhang Y, Zhang C, Feng R, Meng T, Peng W, Song J, Ma W, Xu W, Chen X, Chen J, Liang C. CXCR4 regulates macrophage M1 polarization by altering glycolysis to promote prostate fibrosis. Cell Commun Signal 2024; 22:456. [PMID: 39327570 PMCID: PMC11426013 DOI: 10.1186/s12964-024-01828-y] [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: 06/17/2024] [Accepted: 09/14/2024] [Indexed: 09/28/2024] Open
Abstract
BACKGROUND C-X-C receptor 4(CXCR4) is widely considered to be a highly conserved G protein-coupled receptor, widely involved in the pathophysiological processes in the human body, including fibrosis. However, its role in regulating macrophage-related inflammation in the fibrotic process of prostatitis has not been confirmed. Here, we aim to describe the role of CXCR4 in modulating macrophage M1 polarization through glycolysis in the development of prostatitis fibrosis. METHODS Use inducible experimental chronic prostatitis as a model of prostatic fibrosis. Reduce CXCR4 expression in immortalized bone marrow-derived macrophages using lentivirus. In the fibrotic mouse model, use adenovirus carrying CXCR4 agonists to detect the silencing of CXCR4 and assess the in vivo effects. RESULTS In this study, we demonstrated that reducing CXCR4 expression during LPS treatment of macrophages can alleviate M1 polarization. Silencing CXCR4 can inhibit glycolytic metabolism, enhance mitochondrial function, and promote macrophage transition from M1 to M2. Additionally, in vivo functional experiments using AAV carrying CXCR4 showed that blocking CXCR4 in experimental autoimmune prostatitis (EAP) can alleviate inflammation and experimental prostate fibrosis development. Mechanistically, CXCR4, a chemokine receptor, when silenced, weakens the PI3K/AKT/mTOR pathway as its downstream signal, reducing c-MYC expression. PFKFB3, a key enzyme involved in glucose metabolism, is a target gene of c-MYC, thus impacting macrophage polarization and glycolytic metabolism processes.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Urology, the First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui, People's Republic of China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, People's Republic of China
- Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Anhui Medical University, Hefei, Anhui, People's Republic of China
| | - Chen Zhang
- Department of Urology, the First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui, People's Republic of China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, People's Republic of China
- Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Anhui Medical University, Hefei, Anhui, People's Republic of China
| | - Rui Feng
- Department of Urology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Tong Meng
- Department of Urology, the First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui, People's Republic of China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, People's Republic of China
- Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Anhui Medical University, Hefei, Anhui, People's Republic of China
| | - Wei Peng
- Department of Urology, the First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui, People's Republic of China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, People's Republic of China
- Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Anhui Medical University, Hefei, Anhui, People's Republic of China
| | - Jian Song
- Department of Urology, the First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui, People's Republic of China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, People's Republic of China
- Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Anhui Medical University, Hefei, Anhui, People's Republic of China
| | - Wenming Ma
- Department of Urology, the First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui, People's Republic of China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, People's Republic of China
- Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Anhui Medical University, Hefei, Anhui, People's Republic of China
| | - Wenlong Xu
- Department of Urology, the First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui, People's Republic of China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, People's Republic of China
- Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Anhui Medical University, Hefei, Anhui, People's Republic of China
| | - Xianguo Chen
- Department of Urology, the First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui, People's Republic of China.
- Institute of Urology, Anhui Medical University, Hefei, Anhui, People's Republic of China.
- Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Anhui Medical University, Hefei, Anhui, People's Republic of China.
| | - Jing Chen
- Department of Urology, the First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui, People's Republic of China.
- Institute of Urology, Anhui Medical University, Hefei, Anhui, People's Republic of China.
- Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Anhui Medical University, Hefei, Anhui, People's Republic of China.
| | - Chaozhao Liang
- Department of Urology, the First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui, People's Republic of China.
- Institute of Urology, Anhui Medical University, Hefei, Anhui, People's Republic of China.
- Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Anhui Medical University, Hefei, Anhui, People's Republic of China.
| |
Collapse
|
9
|
Chen HR, Sun Y, Mittler G, Rumpf T, Shvedunova M, Grosschedl R, Akhtar A. MOF-mediated PRDX1 acetylation regulates inflammatory macrophage activation. Cell Rep 2024; 43:114682. [PMID: 39207899 DOI: 10.1016/j.celrep.2024.114682] [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: 10/09/2023] [Revised: 06/27/2024] [Accepted: 08/09/2024] [Indexed: 09/04/2024] Open
Abstract
Signaling-dependent changes in protein phosphorylation are critical to enable coordination of transcription and metabolism during macrophage activation. However, the role of acetylation in signal transduction during macrophage activation remains obscure. Here, we identify the redox signaling regulator peroxiredoxin 1 (PRDX1) as a substrate of the lysine acetyltransferase MOF. MOF acetylates PRDX1 at lysine 197, preventing hyperoxidation and thus maintaining its activity under stress. PRDX1 K197ac responds to inflammatory signals, decreasing rapidly in mouse macrophages stimulated with bacterial lipopolysaccharides (LPSs) but not with interleukin (IL)-4 or IL-10. The LPS-induced decrease of PRDX1 K197ac elevates cellular hydrogen peroxide accumulation and augments ERK1/2, but not p38 or AKT, phosphorylation. Concomitantly, diminished PRDX1 K197ac stimulates glycolysis, potentiates H3 serine 28 phosphorylation, and ultimately enhances the production of pro-inflammatory mediators such as IL-6. Our work reveals a regulatory role for redox protein acetylation in signal transduction and coordinating metabolic and transcriptional programs during inflammatory macrophage activation.
Collapse
Affiliation(s)
- Hui-Ru Chen
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Baden-Württemberg, Germany; Albert-Ludwigs-University Freiburg, Faculty of Biology, Freiburg, Baden-Württemberg, Germany
| | - Yidan Sun
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Baden-Württemberg, Germany
| | - Gerhard Mittler
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Baden-Württemberg, Germany
| | - Tobias Rumpf
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Baden-Württemberg, Germany
| | - Maria Shvedunova
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Baden-Württemberg, Germany
| | - Rudolf Grosschedl
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Baden-Württemberg, Germany
| | - Asifa Akhtar
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Baden-Württemberg, Germany.
| |
Collapse
|
10
|
Agarwal M, Bhaskar A, Singha B, Mukhopadhyay S, Pahuja I, Singh A, Chaturvedi S, Agarwal N, Dwivedi VP, Nandicoori VK. Depletion of essential mycobacterial gene glmM reduces pathogen survival and induces host-protective immune responses against tuberculosis. Commun Biol 2024; 7:949. [PMID: 39107377 PMCID: PMC11303689 DOI: 10.1038/s42003-024-06620-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 07/23/2024] [Indexed: 08/10/2024] Open
Abstract
The limitations of TB treatment are the long duration and immune-dampening effects of anti-tuberculosis therapy. The Cell wall plays a crucial role in survival and virulence; hence, enzymes involved in its biosynthesis are good therapeutic targets. Here, we identify Mycobacterium tuberculosis (Mtb) GlmM, (GlmMMtb) engaged in the UDP-GlcNAc synthesis pathway as an essential enzyme. We generated a conditional knockdown strain, Rv-glmMkD using the CRISPR interference-mediated gene silencing approach. Depletion of GlmMMtb affects the morphology and thickness of the cell wall. The Rv-glmMkD strain attenuated Mtb survival in vitro, in the host macrophages (ex vivo), and in a murine mice infection model (in vivo). Results suggest that the depletion of GlmMMtb induces M1 macrophage polarization, prompting a pro-inflammatory cytokine response, apparent from the upregulation of activation markers, including IFNɣ and IL-17 that resists the growth of Mtb. These observations provide a rationale for exploring GlmMMtb as a potential therapeutic target.
Collapse
Affiliation(s)
- Meetu Agarwal
- Signal Transduction Laboratory, National Institute of Immunology, New Delhi, India.
- Department of Molecular Medicine, Jamia Hamdard University, New Delhi, India.
| | - Ashima Bhaskar
- Immunobiology Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Biplab Singha
- Signal Transduction Laboratory, National Institute of Immunology, New Delhi, India
| | - Suparba Mukhopadhyay
- Immunobiology Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Isha Pahuja
- Immunobiology Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Archna Singh
- CSIR - Institute of Genomics and Integrative Biology, Mall Road, Delhi, India
| | - Shivam Chaturvedi
- Immunobiology Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Nisheeth Agarwal
- Translational Health Science and Technology Institute (THSTI), Faridabad, India
| | - Ved Prakash Dwivedi
- Immunobiology Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Vinay Kumar Nandicoori
- Signal Transduction Laboratory, National Institute of Immunology, New Delhi, India.
- CSIR-Centre for Cellular and Molecular Biology, Habsiguda, Hyderabad, Telangana, India.
| |
Collapse
|
11
|
Yan L, Wang J, Cai X, Liou Y, Shen H, Hao J, Huang C, Luo G, He W. Macrophage plasticity: signaling pathways, tissue repair, and regeneration. MedComm (Beijing) 2024; 5:e658. [PMID: 39092292 PMCID: PMC11292402 DOI: 10.1002/mco2.658] [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: 03/03/2024] [Revised: 06/24/2024] [Accepted: 06/25/2024] [Indexed: 08/04/2024] Open
Abstract
Macrophages are versatile immune cells with remarkable plasticity, enabling them to adapt to diverse tissue microenvironments and perform various functions. Traditionally categorized into classically activated (M1) and alternatively activated (M2) phenotypes, recent advances have revealed a spectrum of macrophage activation states that extend beyond this dichotomy. The complex interplay of signaling pathways, transcriptional regulators, and epigenetic modifications orchestrates macrophage polarization, allowing them to respond to various stimuli dynamically. Here, we provide a comprehensive overview of the signaling cascades governing macrophage plasticity, focusing on the roles of Toll-like receptors, signal transducer and activator of transcription proteins, nuclear receptors, and microRNAs. We also discuss the emerging concepts of macrophage metabolic reprogramming and trained immunity, contributing to their functional adaptability. Macrophage plasticity plays a pivotal role in tissue repair and regeneration, with macrophages coordinating inflammation, angiogenesis, and matrix remodeling to restore tissue homeostasis. By harnessing the potential of macrophage plasticity, novel therapeutic strategies targeting macrophage polarization could be developed for various diseases, including chronic wounds, fibrotic disorders, and inflammatory conditions. Ultimately, a deeper understanding of the molecular mechanisms underpinning macrophage plasticity will pave the way for innovative regenerative medicine and tissue engineering approaches.
Collapse
Affiliation(s)
- Lingfeng Yan
- Institute of Burn ResearchState Key Laboratory of Trauma and Chemical Poisoningthe First Affiliated Hospital of Army Medical University (the Third Military Medical University)ChongqingChina
- Chongqing Key Laboratory for Wound Damage Repair and RegenerationChongqingChina
| | - Jue Wang
- Institute of Burn ResearchState Key Laboratory of Trauma and Chemical Poisoningthe First Affiliated Hospital of Army Medical University (the Third Military Medical University)ChongqingChina
- Chongqing Key Laboratory for Wound Damage Repair and RegenerationChongqingChina
| | - Xin Cai
- Institute of Burn ResearchState Key Laboratory of Trauma and Chemical Poisoningthe First Affiliated Hospital of Army Medical University (the Third Military Medical University)ChongqingChina
- Chongqing Key Laboratory for Wound Damage Repair and RegenerationChongqingChina
| | - Yih‐Cherng Liou
- Department of Biological SciencesFaculty of ScienceNational University of SingaporeSingaporeSingapore
- National University of Singapore (NUS) Graduate School for Integrative Sciences and EngineeringNational University of SingaporeSingaporeSingapore
| | - Han‐Ming Shen
- Faculty of Health SciencesUniversity of MacauMacauChina
| | - Jianlei Hao
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and TreatmentZhuhai Institute of Translational MedicineZhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University)Jinan UniversityZhuhaiGuangdongChina
- The Biomedical Translational Research InstituteFaculty of Medical ScienceJinan UniversityGuangzhouGuangdongChina
| | - Canhua Huang
- State Key Laboratory of Biotherapy and Cancer CenterWest China Hospitaland West China School of Basic Medical Sciences and Forensic MedicineSichuan University, and Collaborative Innovation Center for BiotherapyChengduChina
| | - Gaoxing Luo
- Institute of Burn ResearchState Key Laboratory of Trauma and Chemical Poisoningthe First Affiliated Hospital of Army Medical University (the Third Military Medical University)ChongqingChina
- Chongqing Key Laboratory for Wound Damage Repair and RegenerationChongqingChina
| | - Weifeng He
- Institute of Burn ResearchState Key Laboratory of Trauma and Chemical Poisoningthe First Affiliated Hospital of Army Medical University (the Third Military Medical University)ChongqingChina
- Chongqing Key Laboratory for Wound Damage Repair and RegenerationChongqingChina
| |
Collapse
|
12
|
Krejčová G, Ruphuy G, Šalamúnová P, Sonntag E, Štěpánek F, Bajgar A. Inhibition of mevalonate pathway by macrophage-specific delivery of atorvastatin prevents their pro-inflammatory polarisation. INSECT MOLECULAR BIOLOGY 2024; 33:323-337. [PMID: 38367277 DOI: 10.1111/imb.12900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 01/30/2024] [Indexed: 02/19/2024]
Abstract
Adjustment of the cellular metabolism of pro-inflammatory macrophages is essential for their bactericidal function; however, it underlies the development of many human diseases if induced chronically. Therefore, intervention of macrophage metabolic polarisation has been recognised as a potent strategy for their treatment. Although many small-molecule inhibitors affecting macrophage metabolism have been identified, their in vivo administration requires a tool for macrophage-specific delivery to limit their potential side effects. Here, we establish Drosophila melanogaster as a simple experimental model for in vivo testing of macrophage-specific delivery tools. We found that yeast-derived glucan particles (GPs) are suitable for macrophage-specific delivery of small-molecule inhibitors. Systemic administration of GPs loaded with atorvastatin, the inhibitor of hydroxy-methyl-glutaryl-CoA reductase (Hmgcr), leads to intervention of mevalonate pathway specifically in macrophages, without affecting HMGCR activity in other tissues. Using this tool, we demonstrate that mevalonate pathway is essential for macrophage pro-inflammatory polarisation and individual's survival of infection.
Collapse
Affiliation(s)
- Gabriela Krejčová
- Faculty of Science, Department of Molecular Biology and Genetics, University of South Bohemia, České Budějovice, Czech Republic
- Biology Centre CAS, Institute of Entomology, České Budějovice, Czech Republic
| | - Gabriela Ruphuy
- Department of Chemical Engineering, University of Chemistry and Technology Prague, Prague, Czech Republic
| | - Petra Šalamúnová
- Department of Chemical Engineering, University of Chemistry and Technology Prague, Prague, Czech Republic
| | - Erik Sonntag
- Department of Chemical Engineering, University of Chemistry and Technology Prague, Prague, Czech Republic
| | - František Štěpánek
- Department of Chemical Engineering, University of Chemistry and Technology Prague, Prague, Czech Republic
| | - Adam Bajgar
- Faculty of Science, Department of Molecular Biology and Genetics, University of South Bohemia, České Budějovice, Czech Republic
- Biology Centre CAS, Institute of Entomology, České Budějovice, Czech Republic
| |
Collapse
|
13
|
Zhu Z, Wang M, Lu S, Dai S, Liu J. Role of macrophage polarization in heart failure and traditional Chinese medicine treatment. Front Pharmacol 2024; 15:1434654. [PMID: 39104386 PMCID: PMC11298811 DOI: 10.3389/fphar.2024.1434654] [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: 05/21/2024] [Accepted: 07/01/2024] [Indexed: 08/07/2024] Open
Abstract
Heart failure (HF) has a severe impact on public health development due to high morbidity and mortality and is associated with imbalances in cardiac immunoregulation. Macrophages, a major cell population involved in cardiac immune response and inflammation, are highly heterogeneous and polarized into M1 and M2 types depending on the microenvironment. M1 macrophage releases inflammatory factors and chemokines to activate the immune response and remove harmful substances, while M2 macrophage releases anti-inflammatory factors to inhibit the overactive immune response and promote tissue repair. M1 and M2 restrict each other to maintain cardiac homeostasis. The dynamic balance of M1 and M2 is closely related to the Traditional Chinese Medicine (TCM) yin-yang theory, and the imbalance of yin and yang will result in a pathological state of the organism. Studies have confirmed that TCM produces positive effects on HF by regulating macrophage polarization. This review describes the critical role of macrophage polarization in inflammation, fibrosis, angiogenesis and electrophysiology in the course of HF, as well as the potential mechanism of TCM regulation of macrophage polarization in preventing and treating HF, thereby providing new ideas for clinical treatment and scientific research design of HF.
Collapse
Affiliation(s)
- Zheqin Zhu
- The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Min Wang
- The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Shenghua Lu
- The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Sisi Dai
- Hunan University of Chinese Medicine, Changsha, China
| | - Jianhe Liu
- The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
14
|
Brown ND, Vomhof-DeKrey EE. Focal Adhesion Kinase and Colony Stimulating Factors: Intestinal Homeostasis and Innate Immunity Crosstalk. Cells 2024; 13:1178. [PMID: 39056760 PMCID: PMC11274384 DOI: 10.3390/cells13141178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 07/08/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
Thousands struggle with acute and chronic intestinal injury due to various causes. Epithelial intestinal healing is dependent on phenotypic transitions to a mobile phenotype. Focal adhesion kinase (FAK) is a ubiquitous protein that is essential for cell mobility. This phenotype change is mediated by FAK activation and proves to be a promising target for pharmaceutical intervention. While FAK is crucial for intestinal healing, new evidence connects FAK with innate immunity and the importance it plays in macrophage/monocyte chemotaxis, as well as other intracellular signaling cascades. These cascades play a part in macrophage/monocyte polarization, maturation, and inflammation that is associated with intestinal injury. Colony stimulating factors (CSFs) such as macrophage colony stimulating factor (M-CSF/CSF-1) and granulocyte macrophage colony stimulating factor (GM-CSF/CSF-2) play a critical role in maintaining homeostasis within intestinal mucosa by crosstalk capabilities between macrophages and epithelial cells. The communication between these cells is imperative in orchestrating healing upon injury. Diving deeper into these connections may allow us a greater insight into the role that our immune system plays in healing, as well as a better comprehension of inflammatory diseases of the gut.
Collapse
Affiliation(s)
- Nicholas D. Brown
- Department of Pathology, School of Medicine and the Health Sciences, University of North Dakota, Grand Forks, ND 58203, USA;
| | - Emilie E. Vomhof-DeKrey
- Department of Pathology, School of Medicine and the Health Sciences, University of North Dakota, Grand Forks, ND 58203, USA;
- Department of Surgery, School of Medicine and the Health Sciences, University of North Dakota, Grand Forks, ND 58203, USA
- Department of Biomedical Sciences, School of Medicine and the Health Sciences, University of North Dakota, Grand Forks, ND 58203, USA
| |
Collapse
|
15
|
Kang J, Lee H, Joo J, Song J, Kim H, Kim YH, Park HR. Comparison of genetic and epigenetic profiles of periodontitis according to the presence of type 2 diabetes. MedComm (Beijing) 2024; 5:e620. [PMID: 38903536 PMCID: PMC11187843 DOI: 10.1002/mco2.620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 05/21/2024] [Accepted: 05/23/2024] [Indexed: 06/22/2024] Open
Abstract
Type 2 diabetes mellitus (T2DM) and periodontitis (PD) have intricated connections as chronic inflammatory diseases. While the immune response is a key factor that accounts for their association, the underlying mechanisms remain unclear. To gain a deeper understanding of the connection, we conducted research using a multiomics approach. We generated whole genome and methylation profiling array data from the periodontium of PD patients with DM (PDDM) and without DM to confirm genetic and epigenetic changes. Independent bulk and single-cell RNA sequencing data were employed to verify the expression levels of hypo-methylated genes. We observed a gradual rise in C>T base substitutions and hypomethylation in PD and PDDM patients compared with healthy participants. Furthermore, specific genetic and epigenetic alterations were prominently associated with the Fc-gamma receptor-mediated phagocytosis pathway. The upregulation of these genes was confirmed in both the periodontal tissues of PD patients and the pancreatic tissues of T2DM patients. Through single-cell RNA analysis of peripheral blood mononuclear cells, substantial upregulation of Fc-gamma receptors and related genes was particularly identified in monocytes. Our findings suggest that targeting the Fc-gamma signaling pathway in monocytes holds promise as a potential treatment strategy for managing systemic complications associated with diabetes.
Collapse
Affiliation(s)
- Junho Kang
- Department of ResearchKeimyung University Dongsan Medical CenterDalseo‐guDaeguRepublic of Korea
| | - Hansong Lee
- Department of BioMedical InformaticsMedical Research Institute, Pusan National UniversityYangsan‐siGyeongsangnam‐doRepublic of Korea
| | - Ji‐Young Joo
- Department of PeriodontologySchool of DentistryPusan National UniversityYangsan‐siGyeongsangnam‐doRepublic of Korea
| | - Jae‐Min Song
- Department of Oral and Maxillofacial SurgerySchool of DentistryPusan National UniversityYangsan‐siGyeongsangnam‐doRepublic of Korea
| | - Hyun‐Joo Kim
- Department of PeriodontologyDental and Life Science InstituteSchool of DentistryPusan National UniversityYangsan‐siGyeongsangnam‐doRepublic of Korea
- Department of Periodontology and Dental Research InstitutePusan National University Dental HospitalYangsan‐siGyeongsangnam‐doRepublic of Korea
- Periodontal Disease Signaling Network Research CenterSchool of DentistryPusan National UniversityYangsan‐siGyeongsangnam‐doRepublic of Korea
| | - Yun Hak Kim
- Periodontal Disease Signaling Network Research CenterSchool of DentistryPusan National UniversityYangsan‐siGyeongsangnam‐doRepublic of Korea
- Department of Biomedical Informatics School of MedicinePusan National UniversityYangsan‐siGyeongsangnam‐doRepublic of Korea
- Department of AnatomySchool of MedicinePusan National UniversityYangsan‐siGyeongsangnam‐doRepublic of Korea
| | - Hae Ryoun Park
- Department of Periodontology and Dental Research InstitutePusan National University Dental HospitalYangsan‐siGyeongsangnam‐doRepublic of Korea
- Periodontal Disease Signaling Network Research CenterSchool of DentistryPusan National UniversityYangsan‐siGyeongsangnam‐doRepublic of Korea
- Department of Oral PathologySchool of DentistryPusan National UniversityYangsan‐siGyeongsangnam‐doRepublic of Korea
| |
Collapse
|
16
|
Qiu H, Xiong H, Zheng J, Peng Y, Wang C, Hu Q, Zhao F, Chen K. Sr-Incorporated Bioactive Glass Remodels the Immunological Microenvironment by Enhancing the Mitochondrial Function of Macrophage via the PI3K/AKT/mTOR Signaling Pathway. ACS Biomater Sci Eng 2024; 10:3923-3934. [PMID: 38766805 DOI: 10.1021/acsbiomaterials.4c00228] [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: 05/22/2024]
Abstract
The repair of critical-sized bone defects continues to pose a challenge in clinics. Strontium (Sr), recognized for its function in bone metabolism regulation, has shown potential in bone repair. However, the underlying mechanism through which Sr2+ guided favorable osteogenesis by modulating macrophages remains unclear, limiting their application in the design of bone biomaterials. Herein, Sr-incorporated bioactive glass (SrBG) was synthesized for further investigation. The release of Sr ions enhanced the immunomodulatory properties and osteogenic potential by modulating the polarization of macrophages toward the M2 phenotype. In vivo, a 3D-printed SrBG scaffold was fabricated and showed consistently improved bone regeneration by creating a prohealing immunological microenvironment. RNA sequencing was performed to explore the underlying mechanisms. It was found that Sr ions might enhance the mitochondrial function of macrophage by activating PI3K/AKT/mTOR signaling, thereby favoring osteogenesis. Our findings demonstrate the relationship between the immunomodulatory role of Sr ions and the mitochondrial function of macrophages. By focusing on the mitochondrial function of macrophages, Sr2+-mediated immunomodulation sheds light on the future design of biomaterials for tissue regenerative engineering.
Collapse
Affiliation(s)
- Huanhuan Qiu
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou 510280, China
| | - Huacui Xiong
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou 510280, China
| | - Jiafu Zheng
- Department of Biomedical Engineering, School of Materials Science and Engineering, South China University of Technology, Guangzhou 510641, China
| | - Yuqi Peng
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou 510280, China
| | - Chunhui Wang
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou 510280, China
| | - Qing Hu
- School of Materials Science and Engineering, Jingdezhen Ceramic University, Jingdezhen 333001, China
| | - Fujian Zhao
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou 510280, China
| | - Ke Chen
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou 510280, China
| |
Collapse
|
17
|
Huang H, Zheng S, Wu J, Liang X, Li S, Mao P, He Z, Chen Y, Sun L, Zhao X, Cai A, Wang L, Sheng H, Yao Q, Chen R, Zhao Y, Kou L. Opsonization Inveigles Macrophages Engulfing Carrier-Free Bilirubin/JPH203 Nanoparticles to Suppress Inflammation for Osteoarthritis Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400713. [PMID: 38593402 PMCID: PMC11165524 DOI: 10.1002/advs.202400713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 03/21/2024] [Indexed: 04/11/2024]
Abstract
Osteoarthritis (OA) is a chronic inflammatory disease characterized by cartilage destruction, synovitis, and osteophyte formation. Disease-modifying treatments for OA are currently lacking. Because inflammation mediated by an imbalance of M1/M2 macrophages in the synovial cavities contributes to OA progression, regulating the M1 to M2 polarization of macrophages can be a potential therapeutic strategy. Basing on the inherent immune mechanism and pathological environment of OA, an immunoglobulin G-conjugated bilirubin/JPH203 self-assembled nanoparticle (IgG/BRJ) is developed, and its therapeutic potential for OA is evaluated. After intra-articular administration, IgG conjugation facilitates the recognition and engulfment of nanoparticles by the M1 macrophages. The internalized nanoparticles disassemble in response to the increased oxidative stress, and the released bilirubin (BR) and JPH203 scavenge reactive oxygen species (ROS), inhibit the nuclear factor kappa-B pathway, and suppress the activated mammalian target of rapamycin pathway, result in the repolarization of macrophages and enhance M2/M1 ratios. Suppression of the inflammatory environment by IgG/BRJ promotes cartilage protection and repair in an OA rat model, thereby improving therapeutic outcomes. This strategy of opsonization involving M1 macrophages to engulf carrier-free BR/JPH203 nanoparticles to suppress inflammation for OA therapy holds great potential for OA intervention and treatment.
Collapse
Affiliation(s)
- Huirong Huang
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of PharmacyThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhou325027China
- Key Laboratory of Structural Malformations in Children of Zhejiang ProvinceWenzhou325027China
- School of Pharmaceutical SciencesWenzhou Medical UniversityWenzhou325035China
| | - Shimin Zheng
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of PharmacyThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhou325027China
- Key Laboratory of Structural Malformations in Children of Zhejiang ProvinceWenzhou325027China
| | - Jianing Wu
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of PharmacyThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhou325027China
- Key Laboratory of Structural Malformations in Children of Zhejiang ProvinceWenzhou325027China
| | - Xindan Liang
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of PharmacyThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhou325027China
- Key Laboratory of Structural Malformations in Children of Zhejiang ProvinceWenzhou325027China
- School of Pharmaceutical SciencesWenzhou Medical UniversityWenzhou325035China
| | - Shengjie Li
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of PharmacyThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhou325027China
- Key Laboratory of Structural Malformations in Children of Zhejiang ProvinceWenzhou325027China
- School of Pharmaceutical SciencesWenzhou Medical UniversityWenzhou325035China
| | - Pengfei Mao
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of PharmacyThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhou325027China
- Key Laboratory of Structural Malformations in Children of Zhejiang ProvinceWenzhou325027China
| | - Zhinan He
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of PharmacyThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhou325027China
- Key Laboratory of Structural Malformations in Children of Zhejiang ProvinceWenzhou325027China
- School of Pharmaceutical SciencesWenzhou Medical UniversityWenzhou325035China
| | - Yahui Chen
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of PharmacyThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhou325027China
- Key Laboratory of Structural Malformations in Children of Zhejiang ProvinceWenzhou325027China
- School of Pharmaceutical SciencesWenzhou Medical UniversityWenzhou325035China
| | - Lining Sun
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of PharmacyThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhou325027China
- Key Laboratory of Structural Malformations in Children of Zhejiang ProvinceWenzhou325027China
- School of Pharmaceutical SciencesWenzhou Medical UniversityWenzhou325035China
| | - Xinyu Zhao
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of PharmacyThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhou325027China
- Key Laboratory of Structural Malformations in Children of Zhejiang ProvinceWenzhou325027China
| | - Aimin Cai
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of PharmacyThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhou325027China
- Key Laboratory of Structural Malformations in Children of Zhejiang ProvinceWenzhou325027China
| | - Luhui Wang
- Department of UltrasonographyThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhou325015China
| | - Huixiang Sheng
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of PharmacyThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhou325027China
| | - Qing Yao
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of PharmacyThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhou325027China
- School of Pharmaceutical SciencesWenzhou Medical UniversityWenzhou325035China
| | - Ruijie Chen
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of PharmacyThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhou325027China
- Key Laboratory of Structural Malformations in Children of Zhejiang ProvinceWenzhou325027China
| | - Ying‐Zheng Zhao
- School of Pharmaceutical SciencesWenzhou Medical UniversityWenzhou325035China
| | - Longfa Kou
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of PharmacyThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhou325027China
- Key Laboratory of Structural Malformations in Children of Zhejiang ProvinceWenzhou325027China
| |
Collapse
|
18
|
Su R, Shao Y, Huang M, Liu D, Yu H, Qiu Y. Immunometabolism in cancer: basic mechanisms and new targeting strategy. Cell Death Discov 2024; 10:236. [PMID: 38755125 PMCID: PMC11099033 DOI: 10.1038/s41420-024-02006-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 05/01/2024] [Accepted: 05/02/2024] [Indexed: 05/18/2024] Open
Abstract
Maturing immunometabolic research empowers immune regulation novel approaches. Progressive metabolic adaptation of tumor cells permits a thriving tumor microenvironment (TME) in which immune cells always lose the initial killing capacity, which remains an unsolved dilemma even with the development of immune checkpoint therapies. In recent years, many studies on tumor immunometabolism have been reported. The development of immunometabolism may facilitate anti-tumor immunotherapy from the recurrent crosstalk between metabolism and immunity. Here, we discuss clinical studies of the core signaling pathways of immunometabolism and their inhibitors or agonists, as well as the specific functions of these pathways in regulating immunity and metabolism, and discuss some of the identified immunometabolic checkpoints. Understanding the comprehensive advances in immunometabolism helps to revise the status quo of cancer treatment. An overview of the new landscape of immunometabolism. The PI3K pathway promotes anabolism and inhibits catabolism. The LKB1 pathway inhibits anabolism and promotes catabolism. Overactivation of PI3K/AKT/mTOR pathway and IDO, IL4I1, ACAT, Sirt2, and MTHFD2 promote immunosuppression of TME formation, as evidenced by increased Treg and decreased T-cell proliferation. The LKBI-AMPK pathway promotes the differentiation of naive T cells to effector T cells and memory T cells and promotes anti-tumor immunity in DCs.
Collapse
Affiliation(s)
- Ranran Su
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin, China
| | - Yingying Shao
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin, China
| | - Manru Huang
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin, China
| | - Donghui Liu
- School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Haiyang Yu
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China.
- Haihe Laboratory of Modern Chinese Medicine, Tianjin, China.
| | - Yuling Qiu
- School of Pharmacy, Tianjin Medical University, Tianjin, China.
| |
Collapse
|
19
|
Liu H, Yao M, Ren J. Codonopsis pilosula-derived glycopeptide dCP1 promotes the polarization of tumor-associated macrophage from M2-like to M1 phenotype. Cancer Immunol Immunother 2024; 73:128. [PMID: 38743074 PMCID: PMC11093951 DOI: 10.1007/s00262-024-03694-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 03/28/2024] [Indexed: 05/16/2024]
Abstract
The majority of the immune cell population in the tumor microenvironment (TME) consists of tumor-associated macrophages (TAM), which are the main players in coordinating tumor-associated inflammation. TAM has a high plasticity and is divided into two main phenotypes, pro-inflammatory M1 type and anti-inflammatory M2 type, with tumor-suppressive and tumor-promoting functions, respectively. Considering the beneficial effects of M1 macrophages for anti-tumor and the high plasticity of macrophages, the conversion of M2 TAM to M1 TAM is feasible and positive for tumor treatment. This study sought to evaluate whether the glycopeptide derived from simulated digested Codonopsis pilosula extracts could regulate the polarization of M2-like TAM toward the M1 phenotype and the potential regulatory mechanisms. The results showed that after glycopeptide dCP1 treatment, the mRNA relative expression levels of some M2 phenotype marker genes in M2-like TAM in simulated TME were reduced, and the relative expression levels of M1 phenotype marker genes and inflammatory factor genes were increased. Analysis of RNA-Seq of M2-like TAM after glycopeptide dCP1 intervention showed that the gene sets such as glycolysis, which is associated with macrophage polarization in the M1 phenotype, were significantly up-regulated, whereas those of gene sets such as IL-6-JAK-STAT3 pathway, which is associated with polarization in the M2 phenotype, were significantly down-regulated. Moreover, PCA analysis and Pearson's correlation also indicated that M2-like TAM polarized toward the M1 phenotype at the transcriptional level after treatment with the glycopeptide dCP1. Lipid metabolomics was used to further explore the efficacy of the glycopeptide dCP1 in regulating the polarization of M2-like TAM to the M1 phenotype. It was found that the lipid metabolite profiles in dCP1-treated M2-like TAM showed M1 phenotype macrophage lipid metabolism profiles compared with blank M2-like TAM. Analysis of the key differential lipid metabolites revealed that the interconversion between phosphatidylcholine (PC) and diacylglycerol (DG) metabolites may be the central reaction of the glycopeptide dCP1 in regulating the conversion of M2-like TAM to the M1 phenotype. The above results suggest that the glycopeptide dCP1 has the efficacy to regulate the polarization of M2-like TAM to M1 phenotype in simulated TME.
Collapse
Affiliation(s)
- Hongxu Liu
- School of Food Science and Engineering, South China University of Technology, Guangzhou, 510641, Guangdong, People's Republic of China
| | - Maojin Yao
- State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, People's Republic of China.
| | - Jiaoyan Ren
- School of Food Science and Engineering, South China University of Technology, Guangzhou, 510641, Guangdong, People's Republic of China.
| |
Collapse
|
20
|
Yurakova TR, Gorshkova EA, Nosenko MA, Drutskaya MS. Metabolic Adaptations and Functional Activity of Macrophages in Homeostasis and Inflammation. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:817-838. [PMID: 38880644 DOI: 10.1134/s0006297924050043] [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: 11/27/2023] [Revised: 02/06/2024] [Accepted: 02/08/2024] [Indexed: 06/18/2024]
Abstract
In recent years, the role of cellular metabolism in immunity has come into the focus of many studies. These processes form a basis for the maintenance of tissue integrity and homeostasis, as well as represent an integral part of the immune response, in particular, inflammation. Metabolic adaptations not only ensure energy supply for immune response, but also affect the functions of immune cells by controlling transcriptional and post-transcriptional programs. Studying the immune cell metabolism facilitates the search for new treatment approaches, especially for metabolic disorders. Macrophages, innate immune cells, are characterized by a high functional plasticity and play a key role in homeostasis and inflammation. Depending on the phenotype and origin, they can either perform various regulatory functions or promote inflammation state, thus exacerbating the pathological condition. Furthermore, their adaptations to the tissue-specific microenvironment influence the intensity and type of immune response. The review examines the effect of metabolic reprogramming in macrophages on the functional activity of these cells and their polarization. The role of immunometabolic adaptations of myeloid cells in tissue homeostasis and in various pathological processes in the context of inflammatory and metabolic diseases is specifically discussed. Finally, modulation of the macrophage metabolism-related mechanisms reviewed as a potential therapeutic approach.
Collapse
Affiliation(s)
- Taisiya R Yurakova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia
| | - Ekaterina A Gorshkova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia
| | - Maxim A Nosenko
- Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, D02F306, Ireland
| | - Marina S Drutskaya
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia.
- Division of Immunobiology and Biomedicine, Center of Genetics and Life Sciences, Sirius University of Science and Technology, Federal Territory Sirius, 354340, Russia
| |
Collapse
|
21
|
Rawat SS, Keshri AK, Arora N, Kaur R, Mishra A, Kumar R, Prasad A. Taenia solium cysticerci's extracellular vesicles Attenuate the AKT/mTORC1 pathway for Alleviating DSS-induced colitis in a murine model. J Extracell Vesicles 2024; 13:e12448. [PMID: 38779712 PMCID: PMC11112404 DOI: 10.1002/jev2.12448] [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: 08/21/2023] [Revised: 03/24/2024] [Accepted: 04/22/2024] [Indexed: 05/25/2024] Open
Abstract
The excretory-secretory proteome plays a pivotal role in both intercellular communication during disease progression and immune escape mechanisms of various pathogens including cestode parasites like Taenia solium. The cysticerci of T. solium causes infection in the central nervous system known as neurocysticercosis (NCC), which affects a significant population in developing countries. Extracellular vesicles (EVs) are 30-150-nm-sized particles and constitute a significant part of the secretome. However, the role of EV in NCC pathogenesis remains undetermined. Here, for the first time, we report that EV from T. solium larvae is abundant in metabolites that can negatively regulate PI3K/AKT pathway, efficiently internalized by macrophages to induce AKT and mTOR degradation through auto-lysosomal route with a prominent increase in the ubiquitination of both proteins. This results in less ROS production and diminished bacterial killing capability among EV-treated macrophages. Due to this, both macro-autophagy and caspase-linked apoptosis are upregulated, with a reduction of the autophagy substrate sequestome 1. In summary, we report that T. solium EV from viable cysts attenuates the AKT-mTOR pathway thereby promoting apoptosis in macrophages, and this may exert immunosuppression during an early viable stage of the parasite in NCC, which is primarily asymptomatic. Further investigation on EV-mediated immune suppression revealed that the EV can protect the mice from DSS-induced colitis and improve colon architecture. These findings shed light on the previously unknown role of T. solium EV and the therapeutic role of their immune suppression potential.
Collapse
Affiliation(s)
- Suraj Singh Rawat
- School of Biosciences and BioengineeringIndian Institute of Technology MandiMandiHimachal PradeshIndia
| | - Anand Kumar Keshri
- School of Biosciences and BioengineeringIndian Institute of Technology MandiMandiHimachal PradeshIndia
| | - Naina Arora
- School of Biosciences and BioengineeringIndian Institute of Technology MandiMandiHimachal PradeshIndia
| | - Rimanpreet Kaur
- School of Biosciences and BioengineeringIndian Institute of Technology MandiMandiHimachal PradeshIndia
| | - Amit Mishra
- Cellular and Molecular Neurobiology UnitIndian Institute of Technology JodhpurJodhpurRajasthanIndia
| | - Rajiv Kumar
- CSIR‐Institute of Himalayan Bioresource TechnologyPalampurHimachal PradeshIndia
| | - Amit Prasad
- School of Biosciences and BioengineeringIndian Institute of Technology MandiMandiHimachal PradeshIndia
| |
Collapse
|
22
|
Liang H, Geng S, Wang Y, Fang Q, Xin Y, Li Y. Tumour-derived exosome SNHG17 induced by oestrogen contributes to ovarian cancer progression via the CCL13-CCR2-M2 macrophage axis. J Cell Mol Med 2024; 28:e18315. [PMID: 38680032 PMCID: PMC11056704 DOI: 10.1111/jcmm.18315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 03/28/2024] [Accepted: 04/03/2024] [Indexed: 05/01/2024] Open
Abstract
Oestrogen is known to be strongly associated with ovarian cancer. There was much work to show the importance of lncRNA SNHG17 in ovarian cancer. However, no study has revealed the molecular regulatory mechanism and functional effects between oestrogen and SNHG17 in the development and metastasis of ovarian cancer. In this study, we found that SNHG17 expression was significantly increased in ovarian cancer and positively correlated with oestrogen treatment. Oestrogen could promote M2 macrophage polarization as well as ovarian cancer cells SKOV3 and ES2 cell exosomal SNHG17 expression. When exposure to oestrogen, exosomal SNHG17 promoted ovarian cancer cell proliferation, migration, invasion and epithelial-mesenchymal transition (EMT) in vitro, and tumour growth and lung metastasis in vivo by accelerating M2-like phenotype of macrophages. Mechanically, exosomal SNHG17 could facilitate the release of CCL13 from M2 macrophage via the PI3K-Akt signalling pathway. Moreover, CCL13-CCR2 axis was identified to be involved in ovarian cancer tumour behaviours driven by oestrogen. There results demonstrate a novel mechanism that exosomal SNHG17 exerts an oncogenic effect on ovarian cancer via the CCL13-CCR2-M2 macrophage axis upon oestrogen treatment, of which SNHG17 may be a potential biomarker and therapeutic target for ovarian cancer responded to oestrogen.
Collapse
Affiliation(s)
- Haiyan Liang
- Department of Obstetrics and GynecologyChina‐Japan Friendship HospitalBeijingChina
| | - Shuo Geng
- Department of Obstetrics and GynecologyChina‐Japan Friendship HospitalBeijingChina
| | - Yadong Wang
- Scientific Research DepartmentGeneX Health Co., LtdBeijingChina
| | - Qing Fang
- Institute of Clinical MedicineChina‐Japan Friendship HospitalBeijingChina
| | - Yongfeng Xin
- Department of GynecologyThe People's Hospital of DaLaTeOrdosInner MongoliaChina
| | - Yanqing Li
- Department of GynecologyHebei Provincial Hospital of Traditional Chinese MedicineWuhanHebeiChina
| |
Collapse
|
23
|
Zhu J, Jiang Q, Gao S, Xia Q, Zhang H, Liu B, Zhao R, Jiang H, Li X, Xu A, Zhou H, Xu Z, Yang C. IL20Rb aggravates pulmonary fibrosis through enhancing bone marrow derived profibrotic macrophage activation. Pharmacol Res 2024; 203:107178. [PMID: 38583686 DOI: 10.1016/j.phrs.2024.107178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 04/03/2024] [Accepted: 04/04/2024] [Indexed: 04/09/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) is one of the most fatal chronic interstitial lung diseases with unknown pathogenesis, current treatments cannot truly reverse the progression of the disease. Pulmonary macrophages, especially bone marrow derived pro-fibrotic macrophages, secrete multiple kinds of profibrotic mediators (SPP1, CD206, CD163, IL-10, CCL18…), thus further promote myofibroblast activation and fibrosis procession. IL20Rb is a cell-surface receptor that belongs to IL-20 family. The role of IL20Rb in macrophage activation and pulmonary fibrosis remains unclear. In this study, we established a bleomycin-induced pulmonary fibrosis model, used IL4/13-inducing THP1 cells to induce profibrotic macrophage (M2-like phenotype) polarization models. We found that IL20Rb is upregulated in the progression of pulmonary fibrosis, and its absence can alleviate the progression of pulmonary fibrosis. In addition, we demonstrated that IL20Rb promote the activation of bone marrow derived profibrotic macrophages by regulating the Jak2/Stat3 and Pi3k/Akt signaling pathways. In terms of therapeutic strategy, we used IL20Rb neutralizing antibodies for animal administration, which was found to alleviate the progression of IPF. Our results suggest that IL20Rb plays a profibrotic role by promoting profibrotic macrophage polarization, and IL20Rb may become a potential therapeutic target for IPF. Neutralizing antibodies against IL20Rb may become a potential drug for the clinical treatment of IPF.
Collapse
Affiliation(s)
- Jingyan Zhu
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300000, China; High-throughput Molecular Drug Screening Centre, Tianjin International Joint Academy of Biomedicine, Tianjin 300070, China
| | - Qiuyan Jiang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300000, China
| | - Shaoyan Gao
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300000, China
| | - Qin Xia
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300000, China; High-throughput Molecular Drug Screening Centre, Tianjin International Joint Academy of Biomedicine, Tianjin 300070, China
| | - Huizhe Zhang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300000, China; High-throughput Molecular Drug Screening Centre, Tianjin International Joint Academy of Biomedicine, Tianjin 300070, China
| | - Bowen Liu
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300000, China
| | - Ruixi Zhao
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300000, China
| | - Haixia Jiang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300000, China; High-throughput Molecular Drug Screening Centre, Tianjin International Joint Academy of Biomedicine, Tianjin 300070, China
| | - Xiaohe Li
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300000, China
| | - Aiguo Xu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China.
| | - Honggang Zhou
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300000, China; High-throughput Molecular Drug Screening Centre, Tianjin International Joint Academy of Biomedicine, Tianjin 300070, China.
| | - Zuojun Xu
- Department of Respiratory and Critical Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China.
| | - Cheng Yang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300000, China; High-throughput Molecular Drug Screening Centre, Tianjin International Joint Academy of Biomedicine, Tianjin 300070, China.
| |
Collapse
|
24
|
Yuan Y, Zhang Y, Lu X, Li J, Wang M, Zhang W, Zheng M, Sun Z, Xing Y, Li Y, Qu Y, Jiao Y, Han H, Xie C, Mao T. Novel insights into macrophage immunometabolism in nonalcoholic steatohepatitis. Int Immunopharmacol 2024; 131:111833. [PMID: 38503012 DOI: 10.1016/j.intimp.2024.111833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 02/29/2024] [Accepted: 03/07/2024] [Indexed: 03/21/2024]
Abstract
Nonalcoholic steatohepatitis (NASH), an inflammatory subtype of nonalcoholic fatty liver disease (NAFLD), is characterized by liver steatosis, inflammation, hepatocellular injury and different degrees of fibrosis, and has been becoming the leading cause of liver-related morbidity and mortality worldwide. Unfortunately, the pathogenesis of NASH has not been completely clarified, and there are no approved therapeutic drugs. Recent accumulated evidences have revealed the involvement of macrophage in the regulation of host liver steatosis, inflammation and fibrosis, and different phenotypes of macrophages have different metabolic characteristics. Therefore, targeted regulation of macrophage immunometabolism may contribute to the treatment and prognosis of NASH. In this review, we summarized the current evidences of the role of macrophage immunometabolism in NASH, especially focused on the related function conversion, as well as the strategies to promote its polarization balance in the liver, and hold promise for macrophage immunometabolism-targeted therapies in the treatment of NASH.
Collapse
Affiliation(s)
- Yali Yuan
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, PR China
| | - Ye Zhang
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, PR China
| | - Xinyu Lu
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, PR China
| | - Junxiang Li
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, PR China
| | - Muyuan Wang
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, PR China
| | - Wenji Zhang
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, PR China
| | | | | | - Yunqi Xing
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, PR China
| | - Yitong Li
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, PR China
| | - Yingdi Qu
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, PR China
| | - Yao Jiao
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, PR China
| | - Haixiao Han
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, PR China.
| | - Chune Xie
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, PR China; Shenzhen Bao'an Traditional Chinese Medicine Hospital, Shenzhen, PR China.
| | - Tangyou Mao
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, PR China.
| |
Collapse
|
25
|
Deng Y, Chen Q, Wan C, Sun Y, Huang F, Hu Y, Yang K. Microglia and macrophage metabolism: a regulator of cerebral gliomas. Cell Biosci 2024; 14:49. [PMID: 38632627 PMCID: PMC11022384 DOI: 10.1186/s13578-024-01231-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 04/07/2024] [Indexed: 04/19/2024] Open
Abstract
Reciprocal interactions between the tumor microenvironment (TME) and cancer cells play important roles in tumorigenesis and progression of glioma. Glioma-associated macrophages (GAMs), either of peripheral origin or representing brain-intrinsic microglia, are the majority population of infiltrating immune cells in glioma. GAMs, usually classified into M1 and M2 phenotypes, have remarkable plasticity and regulate tumor progression through different metabolic pathways. Recently, research efforts have increasingly focused on GAMs metabolism as potential targets for glioma therapy. This review aims to delineate the metabolic characteristics of GAMs within the TME and provide a summary of current therapeutic strategies targeting GAMs metabolism in glioma. The goal is to provide novel insights and therapeutic pathways for glioma by highlighting the significance of GAMs metabolism.
Collapse
Affiliation(s)
- Yue Deng
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, 430022, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Qinyan Chen
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, 430022, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Chao Wan
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, 430022, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yajie Sun
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, 430022, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Fang Huang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, 430022, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yan Hu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, 430022, China.
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Kunyu Yang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, 430022, China.
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
26
|
Quan YZ, Ma A, Ren CQ, An YP, Qiao PS, Gao C, Zhang YK, Li XW, Lin SM, Li NN, Chen DL, Pan Y, Zhou H, Lin DM, Lin SQ, Li M, Yang BX. Ganoderic acids alleviate atherosclerosis by inhibiting macrophage M1 polarization via TLR4/MyD88/NF-κB signaling pathway. Atherosclerosis 2024; 391:117478. [PMID: 38417185 DOI: 10.1016/j.atherosclerosis.2024.117478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/01/2024] [Accepted: 02/07/2024] [Indexed: 03/01/2024]
Abstract
BACKGROUND AND AIMS Atherosclerosis (AS) is a chronic inflammatory disease characterized by lipid infiltration and plaque formation in blood vessel walls. Ganoderic acids (GA), a class of major bioactive compounds isolated from the Chinese traditional medicine Ganoderma lucidum, have multiple pharmacological activities. This study aimed to determine the anti-atherosclerotic effect of GA and reveal the pharmacological mechanism. METHODS ApoE-/- mice were fed a high-cholesterol diet and treated with GA for 16 weeks to induce AS and identify the effect of GA. Network pharmacological analysis was performed to predict the anti-atherosclerotic mechanisms. An invitro cell model was used to explore the effect of GA on macrophage polarization and the possible mechanism involved in bone marrow dereived macrophages (BMDMs) and RAW264.7 cells stimulated with lipopolysaccharide or oxidized low-density lipoprotein. RESULTS It was found that GA at 5 and 25 mg/kg/d significantly inhibited the development of AS and increased plaque stability, as evidenced by decreased plaque in the aorta, reduced necrotic core size and increased collagen/lipid ratio in lesions. GA reduced the proportion of M1 macrophages in plaques, but had no effect on M2 macrophages. In vitro experiments showed that GA (1, 5, 25 μg/mL) significantly decreased the proportion of CD86+ macrophages and the mRNA levels of IL-6, IL-1β, and MCP-1 in macrophages. Experimental results showed that GA inhibited M1 macrophage polarization by regulating TLR4/MyD88/NF-κB signaling pathway. CONCLUSIONS This study demonstrated that GA play an important role in plaque stability and macrophage polarization. GA exert the anti-atherosclerotic effect partly by regulating TLR4/MyD88/NF-κB signaling pathways to inhibit M1 polarization of macrophages. Our study provides theoretical basis and experimental data for the pharmacological activity and mechanisms of GA against AS.
Collapse
Affiliation(s)
- Ya-Zhu Quan
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Ang Ma
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China; Artemisinin Research Center, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100007, China
| | - Chao-Qun Ren
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Yong-Pan An
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Pan-Shuang Qiao
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Cai Gao
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Yu-Kun Zhang
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China; Chongqing Key Laboratory of Development and Utilization of Genuine Medicinal Materials in Three Gorges Reservoir Area, Chongqing Three Gorges Medical College, Chongqing, 404020, China
| | - Xiao-Wei Li
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China; China Resources Pharmaceutical Group Limited, Beijing, 100000, China
| | - Si-Mei Lin
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Nan-Nan Li
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Di-Long Chen
- Chongqing Key Laboratory of Development and Utilization of Genuine Medicinal Materials in Three Gorges Reservoir Area, Chongqing Three Gorges Medical College, Chongqing, 404020, China
| | - Yan Pan
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Hong Zhou
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Dong-Mei Lin
- China National Engineering Research Center on JUNCAO Technology, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Shu-Qian Lin
- China National Engineering Research Center on JUNCAO Technology, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Min Li
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Bao-Xue Yang
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China.
| |
Collapse
|
27
|
Arosa L, Camba-Gómez M, Lorenzo-Martín LF, Clavaín L, López M, Conde-Aranda J. RNA Expression of MMP12 Is Strongly Associated with Inflammatory Bowel Disease and Is Regulated by Metabolic Pathways in RAW 264.7 Macrophages. Int J Mol Sci 2024; 25:3167. [PMID: 38542140 PMCID: PMC10970096 DOI: 10.3390/ijms25063167] [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: 02/02/2024] [Revised: 03/06/2024] [Accepted: 03/07/2024] [Indexed: 04/04/2024] Open
Abstract
Macrophage metalloelastase or matrix metalloproteinase-12 (MMP12) is a macrophage-specific proteolytic enzyme involved in the physiopathology of many inflammatory diseases, including inflammatory bowel disease. Although previously published data suggested that the modulation of MMP12 in macrophages could be a determinant for the development of intestinal inflammation, scarce information is available on the mechanisms underlying the regulation of MMP12 expression in those phagocytes. Therefore, in this study, we aimed to delineate the association of MMP12 with inflammatory bowel disease and the molecular events leading to the transcriptional control of this metalloproteinase. For that, we used publicly available transcriptional data. Also, we worked with the RAW 264.7 macrophage cell line for functional experiments. Our results showed a strong association of MMP12 expression with the severity of inflammatory bowel disease and the response to relevant biological therapies. In vitro assays revealed that the inhibition of mechanistic target of rapamycin complex 1 (mTORC1) and the stimulation of the AMP-activated protein kinase (AMPK) signaling pathway potentiated the expression of Mmp12. Additionally, AMPK and mTOR required a functional downstream glycolytic pathway to fully engage with Mmp12 expression. Finally, the pharmacological inhibition of MMP12 abolished the expression of the proinflammatory cytokine Interleukin-6 (Il6) in macrophages. Overall, our findings provide a better understanding of the mechanistic regulation of MMP12 in macrophages and its relationship with inflammation.
Collapse
Affiliation(s)
- Laura Arosa
- Molecular and Cellular Gastroenterology Group, Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (L.A.); (M.C.-G.)
| | - Miguel Camba-Gómez
- Molecular and Cellular Gastroenterology Group, Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (L.A.); (M.C.-G.)
| | | | - Laura Clavaín
- EGO Genomics, Scientific Park of the University of Salamanca, Adaja Street 4, Building M2, 37185 Villamayor, Spain;
| | - Miguel López
- NeurObesity Group, Department of Physiology, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain;
- CIBER Fisiopatologia de la Obesidad y Nutrición (CIBERobn), 15706 Santiago de Compostela, Spain
| | - Javier Conde-Aranda
- Molecular and Cellular Gastroenterology Group, Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (L.A.); (M.C.-G.)
| |
Collapse
|
28
|
Yan H, Liu Y, Li X, Yu B, He J, Mao X, Yu J, Huang Z, Luo Y, Luo J, Wu A, Chen D. Leucine alleviates cytokine storm syndrome by regulating macrophage polarization via the mTORC1/LXRα signaling pathway. eLife 2024; 12:RP89750. [PMID: 38442142 PMCID: PMC10942637 DOI: 10.7554/elife.89750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2024] Open
Abstract
Cytokine storms are associated with severe pathological damage and death in some diseases. Excessive activation of M1 macrophages and the subsequent secretion of pro-inflammatory cytokines are a major cause of cytokine storms. Therefore, promoting the polarization of M2 macrophages to restore immune balance is a promising therapeutic strategy for treating cytokine storm syndrome (CSS). This study was aimed at investigating the potential protective effects of leucine on lipopolysaccharide (LPS)-induced CSS in mice and exploring the underlying mechanisms. CSS was induced by LPS administration in mice, which were concurrently administered leucine orally. In vitro, bone marrow derived macrophages (BMDMs) were polarized to M1 and M2 phenotypes with LPS and interleukin-4 (IL-4), respectively, and treated with leucine. Leucine decreased mortality in mice treated with lethal doses of LPS. Specifically, leucine decreased M1 polarization and promoted M2 polarization, thus diminishing pro-inflammatory cytokine levels and ameliorating CSS in mice. Further studies revealed that leucine-induced macrophage polarization through the mechanistic target of rapamycin complex 1 (mTORC1)/liver X receptor α (LXRα) pathway, which synergistically enhanced the expression of the IL-4-induced M2 marker Arg1 and subsequent M2 polarization. In summary, this study revealed that leucine ameliorates CSS in LPS mice by promoting M2 polarization through the mTORC1/LXRα/Arg1 signaling pathway. Our findings indicate that a fundamental link between metabolism and immunity contributes to the resolution of inflammation and the repair of damaged tissues.
Collapse
Affiliation(s)
- Hui Yan
- Key Laboratory of Animal Disease Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease resistant Nutrition of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural UniversityChengduChina
| | - Yao Liu
- Key Laboratory of Animal Disease Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease resistant Nutrition of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural UniversityChengduChina
| | - Xipeng Li
- Key Laboratory of Animal Disease Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease resistant Nutrition of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural UniversityChengduChina
| | - Bing Yu
- Key Laboratory of Animal Disease Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease resistant Nutrition of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural UniversityChengduChina
| | - Jun He
- Key Laboratory of Animal Disease Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease resistant Nutrition of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural UniversityChengduChina
| | - Xiangbing Mao
- Key Laboratory of Animal Disease Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease resistant Nutrition of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural UniversityChengduChina
| | - Jie Yu
- Key Laboratory of Animal Disease Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease resistant Nutrition of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural UniversityChengduChina
| | - Zhiqing Huang
- Key Laboratory of Animal Disease Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease resistant Nutrition of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural UniversityChengduChina
| | - Yuheng Luo
- Key Laboratory of Animal Disease Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease resistant Nutrition of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural UniversityChengduChina
| | - Junqiu Luo
- Key Laboratory of Animal Disease Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease resistant Nutrition of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural UniversityChengduChina
| | - Aimin Wu
- Key Laboratory of Animal Disease Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease resistant Nutrition of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural UniversityChengduChina
| | - Daiwen Chen
- Key Laboratory of Animal Disease Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease resistant Nutrition of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural UniversityChengduChina
| |
Collapse
|
29
|
Wang J, Zhao X, Wang Q, Zheng X, Simayi D, Zhao J, Yang P, Mao Q, Xia H. FAM76B regulates PI3K/Akt/NF-κB-mediated M1 macrophage polarization by influencing the stability of PIK3CD mRNA. Cell Mol Life Sci 2024; 81:107. [PMID: 38421448 PMCID: PMC10904503 DOI: 10.1007/s00018-024-05133-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 01/17/2024] [Accepted: 01/17/2024] [Indexed: 03/02/2024]
Abstract
Macrophage polarization is closely related to inflammation development, yet how macrophages are polarized remains unclear. In our study, the number of M1 macrophages was markedly increased in Fam76b knockout U937 cells vs. wild-type U937 cells, and FAM76B expression was decreased in M1 macrophages induced from different sources of macrophages. Moreover, Fam76b knockout enhanced the mRNA and protein levels of M1 macrophage-associated marker genes. These results suggest that FAM76B inhibits M1 macrophage polarization. We then further explored the mechanism by which FAM76B regulates macrophage polarization. We found that FAM76B can regulate PI3K/Akt/NF-κB pathway-mediated M1 macrophage polarization by stabilizing PIK3CD mRNA. Finally, FAM76B was proven to protect against inflammatory bowel disease (IBD) by inhibiting M1 macrophage polarization through the PI3K/Akt/NF-κB pathway in vivo. In summary, FAM76B regulates M1 macrophage polarization through the PI3K/Akt/NF-κB pathway in vitro and in vivo, which may inform the development of future therapeutic strategies for IBD and other inflammatory diseases.
Collapse
Affiliation(s)
- Juan Wang
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, 199 South Chang'an Road, Xi'an, 710062, Shaanxi Province, People's Republic of China
- Department of Pathology, School of Basic Medical Science, Ningxia Medical University, Yinchuan, 750004, People's Republic of China
| | - Xinyue Zhao
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, 199 South Chang'an Road, Xi'an, 710062, Shaanxi Province, People's Republic of China
| | - Qizhi Wang
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, 199 South Chang'an Road, Xi'an, 710062, Shaanxi Province, People's Republic of China
| | - Xiaojing Zheng
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, 199 South Chang'an Road, Xi'an, 710062, Shaanxi Province, People's Republic of China
| | - Dilihumaer Simayi
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, 199 South Chang'an Road, Xi'an, 710062, Shaanxi Province, People's Republic of China
| | - Junli Zhao
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, 199 South Chang'an Road, Xi'an, 710062, Shaanxi Province, People's Republic of China
| | - Peiyan Yang
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, 199 South Chang'an Road, Xi'an, 710062, Shaanxi Province, People's Republic of China
| | - Qinwen Mao
- Department of Pathology, University of Utah, Huntsman Cancer Institute, 2000 Circle of Hope Drive, Salt Lake City, UT, 84112, USA
| | - Haibin Xia
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, 199 South Chang'an Road, Xi'an, 710062, Shaanxi Province, People's Republic of China.
| |
Collapse
|
30
|
Xu Y, Tong X, Liu P, Huang J, Chen S, Liu D, Gu T, Xie Y, Guo D, Xu Y. Deficiency of INPP4A promotes M2 macrophage polarization in eosinophilic chronic rhinosinusitis with nasal polyps. Inflamm Res 2024:10.1007/s00011-024-01855-y. [PMID: 38363325 DOI: 10.1007/s00011-024-01855-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 01/19/2024] [Accepted: 01/26/2024] [Indexed: 02/17/2024] Open
Abstract
OBJECTIVE The treatment of eosinophilic chronic rhinosinusitis with nasal polyps (E-CRSwNP) remains a challenge due to its complex pathogenesis. Inositol polyphosphate-4-phosphatase type IA (INPP4A), a lipid phosphatase, has been implicated in allergic asthma. However, the expression and function of INPP4A in E-CRSwNP remain unclear. This study aims to investigate the role of INPP4A in macrophages in E-CRSwNP. METHODS We assessed the expression of INPP4A in human and mouse nasal mucosal tissues via immunofluorescence staining. THP-1 cells were cultured and exposed to various cytokines to investigate the regulation of INPP4A expression and its functional role. Additionally, we established a murine nasal polyp (NP) model and administrated an INPP4A-overexpressing lentivirus evaluate its impact on NP. RESULTS The percentage of INPP4A + CD68 + macrophages among total macrophages decreased in the E-CRSwNP group compared to the control and the non-eosinophilic CRSwNP (NE-CRSwNP) groups, exhibiting an inverse correlation with an increased percentage of CD206 + CD68 + M2 macrophages among total macrophages. Overexpression of INPP4A led to a reduced percentage of THP-1 cells polarizing towards the M2 phenotype, accompanied by decreased levels of associated chemotactic factors including CCL18, CCL22, CCL24, and CCL26. We also validated the involvement of the PI3K-AKT pathway in the function of INPP4A in vitro. Furthermore, INPP4A overexpression in the murine NP model resulted in the attenuation of eosinophilic inflammation in the nasal mucosa. CONCLUSIONS INPP4A deficiency promotes macrophage polarization towards the M2 phenotype, leading to the secretion of chemokines that recruit eosinophils and Th2 cells, thereby amplifying eosinophilic inflammation in E-CRSwNP. INPP4A may exert a suppressive role in eosinophilic inflammation and could potentially serve as a novel therapeutic strategy.
Collapse
Affiliation(s)
- Yingying Xu
- Department of Rhinology and Allergy, Otolaryngology-Head and Neck Surgery Center, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China
| | - Xiaoting Tong
- Department of Rhinology and Allergy, Otolaryngology-Head and Neck Surgery Center, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China
| | - Peiqiang Liu
- Department of Rhinology and Allergy, Otolaryngology-Head and Neck Surgery Center, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China
| | - Jingyu Huang
- Department of Rhinology and Allergy, Otolaryngology-Head and Neck Surgery Center, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China
| | - Siyuan Chen
- Department of Rhinology and Allergy, Otolaryngology-Head and Neck Surgery Center, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China
| | - Duo Liu
- Department of Rhinology and Allergy, Otolaryngology-Head and Neck Surgery Center, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China
| | - Tian Gu
- Department of Rhinology and Allergy, Otolaryngology-Head and Neck Surgery Center, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China
| | - Yulie Xie
- Department of Rhinology and Allergy, Otolaryngology-Head and Neck Surgery Center, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China
| | - Duo Guo
- Department of Rhinology and Allergy, Otolaryngology-Head and Neck Surgery Center, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China
| | - Yu Xu
- Department of Rhinology and Allergy, Otolaryngology-Head and Neck Surgery Center, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China.
- Hubei Province Key Laboratory of Allergy and Immunity, Wuhan, China.
- Research Institute of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
31
|
Zhang G, Song S, Chen Z, Liu X, Zheng J, Wang Y, Chen X, Song Y. Inhibition of PTEN promotes osteointegration of titanium implants in type 2 diabetes by enhancing anti-inflammation and osteogenic capacity of adipose-derived stem cells. Front Bioeng Biotechnol 2024; 12:1358802. [PMID: 38425992 PMCID: PMC10902433 DOI: 10.3389/fbioe.2024.1358802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 02/02/2024] [Indexed: 03/02/2024] Open
Abstract
Background: The low osteogenic differentiation potential and attenuated anti-inflammatory effect of adipose-derived stem cells (ADSCs) from animals with type 2 diabetes mellitus (T2DM) limits osseointegration of the implant. However, the underlying mechanisms are not fully understood. Methods: Western blotting and qRT-PCR analyses were performed to investigate the effects of PTEN on the osteogenic capacity of ADSCs of T2DM rats (TADSCs). We conducted animal experiments in T2DM-Sprague Dawley (SD) rats to evaluate the osteogenic capacity of modified TADSC sheets in vivo. New bone formation was assessed by micro-CT and histological analyses. Results: In this study, adipose-derived stem cells of T2DM rats exhibited an impaired osteogenic capacity. RNA-seq analysis showed that PTEN mRNA expression was upregulated in TADSCs, which attenuated the osteogenic capacity of TADSCs by inhibiting the AKT/mTOR/HIF-1α signaling pathway. miR-140-3p, which inhibits PTEN, was suppressed in TADSCs. Overexpression or inhibition of PTEN could correspondingly reduce or enhance the osteogenic ability of TADSCs by regulating the AKT/mTOR/HIF-1α signaling pathway. TADSCs transfected with PTEN siRNA resulted in higher and lower expressions of genes encoded in M2 macrophages (Arg1) and M1 macrophages (iNOS), respectively. In the T2DM rat model, PTEN inhibition in TADSC sheets promoted macrophage polarization toward the M2 phenotype, attenuated inflammation, and enhanced osseointegration around implants. Conclusion: Upregulation of PTEN, which was partially due to the inhibition of miR-140-3p, is important for the attenuated osteogenesis by TADSCs owing to the inhibition of the AKT/mTOR/HIF-1α signaling pathway. Inhibition of PTEN significantly improves the anti-inflammatory effect and osteogenic capacity of TADSCs, thus promoting peri-implant bone formation in T2DM rats. Our findings offer a potential therapeutic approach for modifying stem cells derived from patients with T2DM to enhance osseointegration.
Collapse
Affiliation(s)
- Guanhua Zhang
- Department of Implant Dentistry, School of Stomatology, State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration and National Clinical Research Center for Oral Diseases and Shaanxi Clinical Research Center for Oral Diseases, The Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Shuang Song
- College of Stomatology, Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi’an Jiaotong University, Xi’an, China
| | - Zijun Chen
- Department of Implant Dentistry, School of Stomatology, State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration and National Clinical Research Center for Oral Diseases and Shaanxi Clinical Research Center for Oral Diseases, The Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Xiangdong Liu
- Department of Implant Dentistry, School of Stomatology, State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration and National Clinical Research Center for Oral Diseases and Shaanxi Clinical Research Center for Oral Diseases, The Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Jian Zheng
- Department of Implant Dentistry, School of Stomatology, State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration and National Clinical Research Center for Oral Diseases and Shaanxi Clinical Research Center for Oral Diseases, The Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Yuxi Wang
- Department of Implant Dentistry, School of Stomatology, State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration and National Clinical Research Center for Oral Diseases and Shaanxi Clinical Research Center for Oral Diseases, The Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Xutao Chen
- Department of Implant Dentistry, School of Stomatology, State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration and National Clinical Research Center for Oral Diseases and Shaanxi Clinical Research Center for Oral Diseases, The Fourth Military Medical University, Xi’an, Shaanxi, China
- Department of Immunology, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Yingliang Song
- Department of Implant Dentistry, School of Stomatology, State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration and National Clinical Research Center for Oral Diseases and Shaanxi Clinical Research Center for Oral Diseases, The Fourth Military Medical University, Xi’an, Shaanxi, China
| |
Collapse
|
32
|
Zhou H, Jiang B, Qian Y, Ke C. The Mechanistic Target of Rapamycin Complex 1 Pathway Contributes to the Anti-Tumor Effect of Granulocyte-Macrophage-Colony-Stimulating Factor-Producing T Helper Cells in Mouse Colorectal Cancer. Immunol Invest 2024; 53:261-280. [PMID: 38050895 DOI: 10.1080/08820139.2023.2290631] [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: 12/07/2023]
Abstract
INTRODUCTION The role of granulocyte-macrophage-colony-stimulating factor-producing T helper (ThGM) cells in colorectal cancer (CRC) development remains unclear. This study characterizes the function of ThGM cells in mouse CRC. METHODS Mouse CRC was induced by administrating azoxymethane and dextran sulfate sodium. The presence of ThGM cells in CRC tissues and the mechanistic target of rapamycin complex 1 (mTORC1) signaling in ThGM cells was detected by flow cytometry. The impact of mTORC1 signaling on ThGM cell function was determined by in vitro culture. The effect of ThGM cells on CRC development was evaluated by adoptive transfer assays. RESULTS ThGM cells, which expressed granulocyte-macrophage-colony-stimulating factor (GM-CSF), accumulated in CRC tissues. mTORC1 signaling is activated in CRC ThGM cells. mTORC1 inhibition by rapamycin suppressed ThGM cell differentiation and proliferation and resulted in the death of differentiating ThGM cells. mTORC1 inhibition in already differentiated ThGM cells did not induce significant cell death but decreased the expression of GM-CSF, interleukin-2, and tumor necrosis factor-alpha while impeding cell proliferation. Furthermore, mTORC1 inhibition diminished the effect of ThGM cells on driving macrophage polarization toward the M1 type, as evidenced by lower expression of pro-inflammatory cytokines, major histocompatibility complex class II molecule, and CD80 in macrophages after co-culture with rapamycin-treated ThGM cells. Lentivirus-mediated knockdown/overexpression of regulatory-associated protein of mTOR (Raptor) confirmed the essential role of mTORC1 in ThGM cell differentiation and function. Adoptively transferred ThGM cells suppressed CRC growth whereas mTORC1 inhibition abolished this effect. CONCLUSION mTORC1 is essential for the anti-CRC activity of ThGM cells.
Collapse
Affiliation(s)
- Hongjian Zhou
- The Department of Gastrointestinal, Hernia and Abdominal Wall Surgery, Wuhan Third Hospital (Tongren Hospital of Wuhan University), Wuhan, Hubei Province, China
| | - Bin Jiang
- The Department of Gastrointestinal, Hernia and Abdominal Wall Surgery, Wuhan Third Hospital (Tongren Hospital of Wuhan University), Wuhan, Hubei Province, China
| | - Yuyuan Qian
- The Department of Gastrointestinal, Hernia and Abdominal Wall Surgery, Wuhan Third Hospital (Tongren Hospital of Wuhan University), Wuhan, Hubei Province, China
| | - Chao Ke
- The Department of Gastrointestinal, Hernia and Abdominal Wall Surgery, Wuhan Third Hospital (Tongren Hospital of Wuhan University), Wuhan, Hubei Province, China
| |
Collapse
|
33
|
Peng Y, Qiao S, Wang H, Shekhar S, Wang S, Yang J, Fan Y, Yang X. Enhancement of Macrophage Immunity against Chlamydial Infection by Natural Killer T Cells. Cells 2024; 13:133. [PMID: 38247825 PMCID: PMC10813948 DOI: 10.3390/cells13020133] [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: 11/15/2023] [Revised: 12/13/2023] [Accepted: 01/08/2024] [Indexed: 01/23/2024] Open
Abstract
Lung macrophage (LM) is vital in host defence against bacterial infections. However, the influence of other innate immune cells on its function, including the polarisation of different subpopulations, remains poorly understood. This study examined the polarisation of LM subpopulations (monocytes/undifferentiated macrophages (Mo/Mφ), interstitial macrophages (IM), and alveolar macrophages (AM)). We further assessed the effect of invariant natural killer T cells (iNKT) on LM polarisation in a protective function against Chlamydia muridarum, an obligate intracellular bacterium, and respiratory tract infection. We found a preferentially increased local Mo/Mφ and IMs with a significant shift to a type-1 macrophage (M1) phenotype and higher expression of iNOS and TNF-α. Interestingly, during the same infection, the alteration of macrophage subpopulations and the shift towards M1 was much less in iNKT KO mice. More importantly, functional testing by adoptively transferring LMs isolated from iNKT KO mice (iNKT KO-Mφ) conferred less protection than those isolated from wild-type mice (WT-Mφ). Further analyses showed significantly reduced gene expression of the JAK/STAT signalling pathway molecules in iNKT KO-Mφ. The data show an important role of iNKT in promoting LM polarisation to the M1 direction, which is functionally relevant to host defence against a human intracellular bacterial infection. The alteration of JAK/STAT signalling molecule gene expression in iNKT KO-Mφ suggests the modulating effect of iNKT is likely through the JAK/STAT pathway.
Collapse
Affiliation(s)
- Ying Peng
- Department of Immunology, Rady Max College of Medicine, University of Manitoba, Winnipeg, MB R3E 0T5, Canada
| | - Sai Qiao
- Department of Immunology, Rady Max College of Medicine, University of Manitoba, Winnipeg, MB R3E 0T5, Canada
| | - Hong Wang
- Department of Immunology, Rady Max College of Medicine, University of Manitoba, Winnipeg, MB R3E 0T5, Canada
- Department of Medical Microbiology, School of Medicine, Shandong University, Jinan 250100, China
| | - Sudhanshu Shekhar
- Department of Immunology, Rady Max College of Medicine, University of Manitoba, Winnipeg, MB R3E 0T5, Canada
| | - Shuhe Wang
- Department of Immunology, Rady Max College of Medicine, University of Manitoba, Winnipeg, MB R3E 0T5, Canada
| | - Jie Yang
- Department of Immunology, Rady Max College of Medicine, University of Manitoba, Winnipeg, MB R3E 0T5, Canada
| | - Yijun Fan
- Department of Immunology, Rady Max College of Medicine, University of Manitoba, Winnipeg, MB R3E 0T5, Canada
| | - Xi Yang
- Department of Immunology, Rady Max College of Medicine, University of Manitoba, Winnipeg, MB R3E 0T5, Canada
| |
Collapse
|
34
|
Zhu J, Zhou T, Menggen M, Aimulajiang K, Wen H. Ghrelin regulating liver activity and its potential effects on liver fibrosis and Echinococcosis. Front Cell Infect Microbiol 2024; 13:1324134. [PMID: 38259969 PMCID: PMC10800934 DOI: 10.3389/fcimb.2023.1324134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 12/15/2023] [Indexed: 01/24/2024] Open
Abstract
Ghrelin widely exists in the central nervous system and peripheral organs, and has biological activities such as maintaining energy homeostasis, regulating lipid metabolism, cell proliferation, immune response, gastrointestinal physiological activities, cognition, memory, circadian rhythm and reward effects. In many benign liver diseases, it may play a hepatoprotective role against steatosis, chronic inflammation, oxidative stress, mitochondrial dysfunction, endoplasmic reticulum stress and apoptosis, and improve liver cell autophagy and immune response to improve disease progression. However, the role of Ghrelin in liver Echinococcosis is currently unclear. This review systematically summarizes the molecular mechanisms by which Ghrelin regulates liver growth metabolism, immune-inflammation, fibrogenesis, proliferation and apoptosis, as well as its protective effects in liver fibrosis diseases, and further proposes the role of Ghrelin in liver Echinococcosis infection. During the infectious process, it may promote the parasitism and survival of parasites on the host by improving the immune-inflammatory microenvironment and fibrosis state, thereby accelerating disease progression. However, there is currently a lack of targeted in vitro and in vivo experimental evidence for this viewpoint.
Collapse
Affiliation(s)
- Jiang Zhu
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
- Department of Hepatobiliary and Hydatid Disease, Digestive and Vascular Surgery Center Therapy Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Tanfang Zhou
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
- Department of Hepatobiliary and Hydatid Disease, Digestive and Vascular Surgery Center Therapy Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Meng Menggen
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Kalibixiati Aimulajiang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Hao Wen
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
- Department of Hepatobiliary and Hydatid Disease, Digestive and Vascular Surgery Center Therapy Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| |
Collapse
|
35
|
Sun D, Chang Q, Lu F. Immunomodulation in diabetic wounds healing: The intersection of macrophage reprogramming and immunotherapeutic hydrogels. J Tissue Eng 2024; 15:20417314241265202. [PMID: 39071896 PMCID: PMC11283672 DOI: 10.1177/20417314241265202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 06/12/2024] [Indexed: 07/30/2024] Open
Abstract
Diabetic wound healing presents a significant clinical challenge due to the interplay of systemic metabolic disturbances and local inflammation, which hinder the healing process. Macrophages undergo a phenotypic shift from M1 to M2 during wound healing, a transition pivotal for effective tissue repair. However, in diabetic wounds, the microenvironment disrupts this phenotypic polarization, perpetuating inflammation, and impeding healing. Reprograming macrophages to restore their M2 phenotype offers a potential avenue for modulating the wound immune microenvironment and promoting healing. This review elucidates the mechanisms underlying impaired macrophage polarization toward the M2 phenotype in diabetic wounds and discusses novel strategies, including epigenetic and metabolic interventions, to promote macrophage conversion to M2. Hydrogels, with their hydrated 3D cross-linked structure, closely resemble the physiological extracellular matrix and offer advantageous properties such as biocompatibility, tunability, and versatility. These characteristics make hydrogels promising candidates for developing immunomodulatory materials aimed at addressing diabetic wounds. Understanding the role of hydrogels in immunotherapy, particularly in the context of macrophage reprograming, is essential for the development of advanced wound care solutions. This review also highlights recent advancements in immunotherapeutic hydrogels as a step toward precise and effective treatments for diabetic wounds.
Collapse
Affiliation(s)
- Dan Sun
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qiang Chang
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Feng Lu
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
36
|
Kim DM, Lee JH, Pan Q, Han HW, Shen Z, Eshghjoo S, Wu CS, Yang W, Noh JY, Threadgill DW, Guo S, Wright G, Alaniz R, Sun Y. Nutrient-sensing growth hormone secretagogue receptor in macrophage programming and meta-inflammation. Mol Metab 2024; 79:101852. [PMID: 38092245 PMCID: PMC10772824 DOI: 10.1016/j.molmet.2023.101852] [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: 10/11/2023] [Revised: 12/03/2023] [Accepted: 12/08/2023] [Indexed: 12/20/2023] Open
Abstract
OBJECTIVE Obesity-associated chronic inflammation, aka meta-inflammation, is a key pathogenic driver for obesity-associated comorbidity. Growth hormone secretagogue receptor (GHSR) is known to mediate the effects of nutrient-sensing hormone ghrelin in food intake and fat deposition. We previously reported that global Ghsr ablation protects against diet-induced inflammation and insulin resistance, but the site(s) of action and mechanism are unknown. Macrophages are key drivers of meta-inflammation. To unravel the role of GHSR in macrophages, we generated myeloid-specific Ghsr knockout mice (LysM-Cre;Ghsrf/f). METHODS LysM-Cre;Ghsrf/f and control Ghsrf/f mice were subjected to 5 months of high-fat diet (HFD) feeding to induce obesity. In vivo, metabolic profiling of food intake, physical activity, and energy expenditure, as well as glucose and insulin tolerance tests (GTT and ITT) were performed. At termination, peritoneal macrophages (PMs), epididymal white adipose tissue (eWAT), and liver were analyzed by flow cytometry and histology. For ex vivo studies, bone marrow-derived macrophages (BMDMs) were generated from the mice and treated with palmitic acid (PA) or lipopolysaccharide (LPS). For in vitro studies, macrophage RAW264.7 cells with Ghsr overexpression or Insulin receptor substrate 2 (Irs2) knockdown were studied. RESULTS We found that Ghsr expression in PMs was increased under HFD feeding. In vivo, HFD-fed LysM-Cre;Ghsrf/f mice exhibited significantly attenuated systemic inflammation and insulin resistance without affecting food intake or body weight. Tissue analysis showed that HFD-fed LysM-Cre;Ghsrf/f mice have significantly decreased monocyte/macrophage infiltration, pro-inflammatory activation, and lipid accumulation, showing elevated lipid-associated macrophages (LAMs) in eWAT and liver. Ex vivo, Ghsr-deficient macrophages protected against PA- or LPS-induced pro-inflammatory polarization, showing reduced glycolysis, increased fatty acid oxidation, and decreased NF-κB nuclear translocation. At molecular level, GHSR metabolically programs macrophage polarization through PKA-CREB-IRS2-AKT2 signaling pathway. CONCLUSIONS These novel results demonstrate that macrophage GHSR plays a key role in the pathogenesis of meta-inflammation, and macrophage GHSR promotes macrophage infiltration and induces pro-inflammatory polarization. These exciting findings suggest that GHSR may serve as a novel immunotherapeutic target for the treatment of obesity and its associated comorbidity.
Collapse
Affiliation(s)
- Da Mi Kim
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA
| | - Jong Han Lee
- Department of Marine Bioindustry, Hanseo University, Seosan 31962, South Korea; USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College Medicine, Houston, TX 77030, USA
| | - Quan Pan
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA
| | - Hye Won Han
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA
| | - Zheng Shen
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA
| | - Sahar Eshghjoo
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX 77807, USA; Agilent technologies, Aanta Clara, CA 95051, USA
| | - Chia-Shan Wu
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA; USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College Medicine, Houston, TX 77030, USA
| | - Wanbao Yang
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA
| | - Ji Yeon Noh
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA
| | - David W Threadgill
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA; Texas A&M Institute for Genome Sciences and Society, Department of Cell Biology and Genetics, Texas A&M University, College Station, TX 77843, USA
| | - Shaodong Guo
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA
| | - Gus Wright
- Department of Veterinary Pathobiology, Texas A&M University, College Station, TX 77843, USA
| | - Robert Alaniz
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX 77807, USA; Tlaloc Therapeutics Inc., College Station, TX 77845, USA
| | - Yuxiang Sun
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA; USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College Medicine, Houston, TX 77030, USA.
| |
Collapse
|
37
|
Xie Y, Zhao G, Lei X, Cui N, Wang H. Advances in the regulatory mechanisms of mTOR in necroptosis. Front Immunol 2023; 14:1297408. [PMID: 38164133 PMCID: PMC10757967 DOI: 10.3389/fimmu.2023.1297408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 12/01/2023] [Indexed: 01/03/2024] Open
Abstract
The mammalian target of rapamycin (mTOR), an evolutionarily highly conserved serine/threonine protein kinase, plays a prominent role in controlling gene expression, metabolism, and cell death. Programmed cell death (PCD) is indispensable for maintaining homeostasis by removing senescent, defective, or malignant cells. Necroptosis, a type of PCD, relies on the interplay between receptor-interacting serine-threonine kinases (RIPKs) and the membrane perforation by mixed lineage kinase domain-like protein (MLKL), which is distinguished from apoptosis. With the development of necroptosis-regulating mechanisms, the importance of mTOR in the complex network of intersecting signaling pathways that govern the process has become more evident. mTOR is directly responsible for the regulation of RIPKs. Autophagy is an indirect mechanism by which mTOR regulates the removal and interaction of RIPKs. Another necroptosis trigger is reactive oxygen species (ROS) produced by oxidative stress; mTOR regulates necroptosis by exploiting ROS. Considering the intricacy of the signal network, it is reasonable to assume that mTOR exerts a bifacial effect on necroptosis. However, additional research is necessary to elucidate the underlying mechanisms. In this review, we summarized the mechanisms underlying mTOR activation and necroptosis and highlighted the signaling pathway through which mTOR regulates necroptosis. The development of therapeutic targets for various diseases has been greatly advanced by the expanding knowledge of how mTOR regulates necroptosis.
Collapse
Affiliation(s)
- Yawen Xie
- Department of Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Guoyu Zhao
- Department of Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Xianli Lei
- Department of Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Na Cui
- Department of Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Hao Wang
- Department of Critical Care Medicine, Beijing Jishuitan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
38
|
Wang J, Tian F, Cao L, Du R, Tong J, Ding X, Yuan Y, Wang C. Macrophage polarization in spinal cord injury repair and the possible role of microRNAs: A review. Heliyon 2023; 9:e22914. [PMID: 38125535 PMCID: PMC10731087 DOI: 10.1016/j.heliyon.2023.e22914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 11/17/2023] [Accepted: 11/22/2023] [Indexed: 12/23/2023] Open
Abstract
The prevention, treatment, and rehabilitation of spinal cord injury (SCI) have always posed significant medical challenges. After mechanical injury, disturbances in microcirculation, edema formation, and the generation of free radicals lead to additional damage, impeding effective repair processes and potentially exacerbating further dysfunction. In this context, inflammatory responses, especially the activation of macrophages, play a pivotal role. Different phenotypes of macrophages have distinct effects on inflammation. Activation of classical macrophage cells (M1) promotes inflammation, while activation of alternative macrophage cells (M2) inhibits inflammation. The polarization of macrophages is crucial for disease healing. A non-coding RNA, known as microRNA (miRNA), governs the polarization of macrophages, thereby reducing inflammation following SCI and facilitating functional recovery. This study elucidates the inflammatory response to SCI, focusing on the infiltration of immune cells, specifically macrophages. It examines their phenotype and provides an explanation of their polarization mechanisms. Finally, this paper introduces several well-known miRNAs that contribute to macrophage polarization following SCI, including miR-155, miR-130a, and miR-27 for M1 polarization, as well as miR-22, miR-146a, miR-21, miR-124, miR-223, miR-93, miR-132, and miR-34a for M2 polarization. The emphasis is placed on their potential therapeutic role in SCI by modulating macrophage polarization, as well as the present developments and obstacles of miRNA clinical therapy.
Collapse
Affiliation(s)
- Jiawei Wang
- School and Hospital of Stomatology, Shanxi Medical University, Shanxi Taiyuan, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Shanxi Taiyuan, China
| | - Feng Tian
- School and Hospital of Stomatology, Shanxi Medical University, Shanxi Taiyuan, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Shanxi Taiyuan, China
| | - Lili Cao
- School and Hospital of Stomatology, Shanxi Medical University, Shanxi Taiyuan, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Shanxi Taiyuan, China
| | - Ruochen Du
- Experimental Animal Center, Shanxi Medical University, Shanxi Taiyuan, China
| | - Jiahui Tong
- School and Hospital of Stomatology, Shanxi Medical University, Shanxi Taiyuan, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Shanxi Taiyuan, China
| | - Xueting Ding
- Experimental Animal Center, Shanxi Medical University, Shanxi Taiyuan, China
| | - Yitong Yuan
- Experimental Animal Center, Shanxi Medical University, Shanxi Taiyuan, China
| | - Chunfang Wang
- School and Hospital of Stomatology, Shanxi Medical University, Shanxi Taiyuan, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Shanxi Taiyuan, China
| |
Collapse
|
39
|
Zhang Q, Chen S, Guo Y, He F, Fu J, Ren W. Phenylalanine diminishes M1 macrophage inflammation. SCIENCE CHINA. LIFE SCIENCES 2023; 66:2862-2876. [PMID: 37243947 DOI: 10.1007/s11427-022-2296-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 02/17/2023] [Indexed: 05/29/2023]
Abstract
Emerging evidence suggests that amino acids dictate the effector functions of immune cells; however, whether and how phenylalanine (Phe) orchestrates the polarization of macrophages is not understood. Here, we determined that Phe attenuated lipopolysaccharide (LPS) and P. multocida serotype A strain CQ2 (PmCQ2) infection-induced inflammation in vivo. Furthermore, we demonstrated that Phe inhibited the production of interleukin (IL)-1β and tumor necrosis factor (TNF)-α in proinflammatory (M1) macrophages. Phe reprogrammed the transcriptomic and metabolic profiles and enhanced oxidative phosphorylation in M1 macrophages, which reduced the activation of caspase-1. Notably, the valine-succinyl-CoA axis played a critical role in Phe-mediated inhibition of IL-1β production in M1 macrophages. Taken together, our findings suggest that manipulating the valine-succinyl-CoA axis provides a potential target for preventing and/or treating macrophage-related diseases.
Collapse
Affiliation(s)
- Qingzhuo Zhang
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Siyuan Chen
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Yan Guo
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Fang He
- College of Animal Science and Technology, Southwest University, Chongqing, 400715, China
| | - Jian Fu
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Wenkai Ren
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China.
| |
Collapse
|
40
|
Raien A, Davis S, Zhang M, Zitser D, Lin M, Pitcher G, Bhalodia K, Subbian S, Venketaraman V. Effects of Everolimus in Modulating the Host Immune Responses against Mycobacterium tuberculosis Infection. Cells 2023; 12:2653. [PMID: 37998388 PMCID: PMC10670413 DOI: 10.3390/cells12222653] [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: 10/09/2023] [Revised: 11/16/2023] [Accepted: 11/17/2023] [Indexed: 11/25/2023] Open
Abstract
The phosphoinositide 3-kinase/protein kinase B/mammalian target of rapamycin (P13K/AKT/mTOR) pathway plays a key role in tuberculosis (TB) pathogenesis and infection. While the activity levels of this pathway during active infection are still debated, manipulating this pathway shows potential benefit for host-directed therapies. Some studies indicate that pathway inhibitors may have potential for TB treatment through upregulation of autophagy, while other studies do not encourage the use of these inhibitors due to possible host tissue destruction by Mycobacterium tuberculosis (M. tb) and increased infection risk. Investigating further clinical trials and their use of pathway inhibitors is necessary in order to ascertain their potential for TB treatment. This paper is particularly focused on the drug everolimus, an mTOR inhibitor. One of the first clinical trials sponsored by the Aurum Institute showed potential benefit in using everolimus as an adjunctive therapy for tuberculosis. Infection with tuberculosis is associated with a metabolic shift from oxidative phosphorylation towards glycolysis. The everolimus arm in the clinical trial showed further reduction than the control for both maximal and peak glycolytic activity. Compared with control, those receiving everolimus demonstrated increased lung function through forced expiratory volume in 1 s (FEV1) measurements, suggesting that everolimus may mitigate inflammation contributing to lung damage.
Collapse
Affiliation(s)
- Anmol Raien
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA; (A.R.); (S.D.); (M.Z.); (D.Z.); (M.L.); (G.P.); (K.B.)
| | - Sofia Davis
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA; (A.R.); (S.D.); (M.Z.); (D.Z.); (M.L.); (G.P.); (K.B.)
| | - Michelle Zhang
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA; (A.R.); (S.D.); (M.Z.); (D.Z.); (M.L.); (G.P.); (K.B.)
| | - David Zitser
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA; (A.R.); (S.D.); (M.Z.); (D.Z.); (M.L.); (G.P.); (K.B.)
| | - Michelle Lin
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA; (A.R.); (S.D.); (M.Z.); (D.Z.); (M.L.); (G.P.); (K.B.)
| | - Graysen Pitcher
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA; (A.R.); (S.D.); (M.Z.); (D.Z.); (M.L.); (G.P.); (K.B.)
| | - Krishna Bhalodia
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA; (A.R.); (S.D.); (M.Z.); (D.Z.); (M.L.); (G.P.); (K.B.)
| | - Selvakumar Subbian
- Public Health Research Center, New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA;
| | - Vishwanath Venketaraman
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA; (A.R.); (S.D.); (M.Z.); (D.Z.); (M.L.); (G.P.); (K.B.)
| |
Collapse
|
41
|
Yan L, Hou C, Liu J, Wang Y, Zeng C, Yu J, Zhou T, Zhou Q, Duan S, Xiong W. Local administration of liposomal-based Plekhf1 gene therapy attenuates pulmonary fibrosis by modulating macrophage polarization. SCIENCE CHINA. LIFE SCIENCES 2023; 66:2571-2586. [PMID: 37340175 DOI: 10.1007/s11427-022-2314-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 03/01/2023] [Indexed: 06/22/2023]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a fatal interstitial lung disease with limited therapeutic options. Macrophages, particularly alternatively activated macrophages (M2), have been recognized to contribute to the pathogenesis of pulmonary fibrosis. Therefore, targeting macrophages might be a viable therapeutic strategy for IPF. Herein, we report a potential nanomedicine-based gene therapy for IPF by modulating macrophage M2 activation. In this study, we illustrated that the levels of pleckstrin homology and FYVE domain containing 1 (Plekhf1) were increased in the lungs originating from IPF patients and PF mice. Further functionality studies identified the pivotal role of Plekhf1 in macrophage M2 activation. Mechanistically, Plekhf1 was upregulated by IL-4/IL-13 stimulation, after which Plekhf1 enhanced PI3K/Akt signaling to promote the macrophage M2 program and exacerbate pulmonary fibrosis. Therefore, intratracheal administration of Plekhf1 siRNA-loaded liposomes could effectively suppress the expression of Plekhf1 in the lungs and notably protect mice against BLM-induced lung injury and fibrosis, concomitant with a significant reduction in M2 macrophage accumulation in the lungs. In conclusion, Plekhf1 may play a crucial role in the pathogenesis of pulmonary fibrosis, and Plekhf1 siRNA-loaded liposomes might be a promising therapeutic approach against pulmonary fibrosis.
Collapse
Affiliation(s)
- Lifeng Yan
- Department of Respiratory and Critical Care Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Chenchen Hou
- Department of Respiratory and Critical Care Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Juan Liu
- Department of Respiratory and Critical Care Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Yi Wang
- Department of Pulmonary and Critical Care Medicine, The Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Chenxi Zeng
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, 430030, China
| | - Jun Yu
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, 430030, China
| | - Tianyu Zhou
- Department of Respiratory and Critical Care Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- Department of Pulmonary and Critical Care Medicine, The Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Qing Zhou
- Department of Pulmonary and Critical Care Medicine, The Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Shengzhong Duan
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, 200011, China.
| | - Weining Xiong
- Department of Respiratory and Critical Care Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
- Shanghai Key Laboratory of Tissue Engineering, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
| |
Collapse
|
42
|
Wang H, Yu H, Huang T, Wang B, Xiang L. Hippo-YAP/TAZ signaling in osteogenesis and macrophage polarization: Therapeutic implications in bone defect repair. Genes Dis 2023; 10:2528-2539. [PMID: 37554194 PMCID: PMC10404961 DOI: 10.1016/j.gendis.2022.12.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 11/16/2022] [Accepted: 12/08/2022] [Indexed: 01/18/2023] Open
Abstract
Bone defects caused by diseases or surgery are a common clinical problem. Researchers are devoted to finding biological mechanisms that accelerate bone defect repair, which is a complex and continuous process controlled by many factors. As members of transcriptional costimulatory molecules, Yes-associated protein (YAP) and transcriptional co-activator with PDZ-binding motif (TAZ) play an important regulatory role in osteogenesis, and they affect cell function by regulating the expression of osteogenic genes in osteogenesis-related cells. Macrophages are an important group of cells whose function is regulated by YAP/TAZ. Currently, the relationship between YAP/TAZ and macrophage polarization has attracted increasing attention. In bone tissue, YAP/TAZ can realize diverse osteogenic regulation by mediating macrophage polarization. Macrophages polarize into M1 and M2 phenotypes under different stimuli. M1 macrophages dominate the inflammatory response by releasing a number of inflammatory mediators in the early phase of bone defect repair, while massive aggregation of M2 macrophages is beneficial for inflammation resolution and tissue repair, as they secrete many anti-inflammatory and osteogenesis-related cytokines. The mechanism of YAP/TAZ-mediated macrophage polarization during osteogenesis warrants further study and it is likely to be a promising strategy for bone defect repair. In this article, we review the effect of Hippo-YAP/TAZ signaling and macrophage polarization on bone defect repair, and highlight the regulation of macrophage polarization by YAP/TAZ.
Collapse
Affiliation(s)
- Haochen Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Hui Yu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Tianyu Huang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Bin Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Lin Xiang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
43
|
Kim YJ, Jin J, Kim DH, Kim D, Lee YM, Byun JK, Choi YK, Park KG. SGLT2 inhibitors prevent LPS-induced M1 macrophage polarization and alleviate inflammatory bowel disease by downregulating NHE1 expression. Inflamm Res 2023; 72:1981-1997. [PMID: 37770568 DOI: 10.1007/s00011-023-01796-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 09/01/2023] [Accepted: 09/09/2023] [Indexed: 09/30/2023] Open
Abstract
BACKGROUND Classically activated M1 macrophages, characterized by aberrant glycolysis and secretion of inflammatory cytokines, play pivotal roles in inflammatory diseases, including inflammatory bowel disease (IBD). Recently, sodium-glucose co-transporter 2 (SGLT2) inhibitors were shown to suppress Na+/H+ exchanger 1 (NHE1) and Na+/Ca2+ exchanger 1 (NCX1) activity, regulating downstream intracellular Ca2+ concentrations in cardiomyocytes. However, whether SGLT2 inhibitors regulate M1 macrophage polarization by downregulating NHE1 and NCX1 remains unknown. METHODS We analyzed cellular responses to SGLT2 inhibitors using mouse bone marrow-derived macrophages and peritoneal macrophages treated with lipopolysaccharide (LPS). To induce IBD, we used a dextran sulfate sodium salt-induced colitis mouse model. RESULTS We observed that NHE1 and NCX1 were overexpressed in LPS-treated macrophages, leading to M1 macrophage polarization. Mechanistically, NHE1 and NCX1-mediated Ca2+ accumulation in the macrophage resulted in enhanced glycolysis by promoting PI3K/AKT/mTORC1 signaling. SGLT2 inhibitors suppressed both the expression levels and activities of NHE1 and NCX1, and consequently downregulated PI3K/AKT/mTORC1 signaling and glycolysis in LPS-treated macrophages. We observed inhibition of LPS-stimulated M1 polarization and cytokine production by SGLT2 inhibitors in vitro, ex vivo, and in an IBD mouse model. CONCLUSIONS NHE1 promotes M1 macrophage polarization and SGLT2 inhibitors are a novel strategy to treat M1 macrophage-mediated inflammatory diseases, including IBD.
Collapse
Affiliation(s)
- Ye Jin Kim
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, 130 Dongdeok-Ro, Jung-Gu, Daegu, 41944, Republic of Korea
- Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Jonghwa Jin
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, 130 Dongdeok-Ro, Jung-Gu, Daegu, 41944, Republic of Korea
| | - Dong-Ho Kim
- Department of Biomedical Science, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Daehoon Kim
- Department of Biomedical Science, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - You Mie Lee
- College of Pharmacy, Vessel-Organ Interaction Research Center (VOICE, MRC), Kyungpook National University, Daegu, 41566, Republic of Korea
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Jun-Kyu Byun
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Yeon-Kyung Choi
- Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, 41566, Republic of Korea.
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, 807 Hoguk-Ro, Buk-Gu, Daegu, 41404, Republic of Korea.
| | - Keun-Gyu Park
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, 130 Dongdeok-Ro, Jung-Gu, Daegu, 41944, Republic of Korea.
- Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, 41566, Republic of Korea.
- Department of Biomedical Science, Kyungpook National University, Daegu, 41566, Republic of Korea.
| |
Collapse
|
44
|
Zhong W, Lu Y, Han X, Yang J, Qin Z, Zhang W, Yu Z, Wu B, Liu S, Xu W, Zheng C, Schuchter LM, Karakousis GC, Mitchell TC, Amaravadi R, Flowers AJ, Gimotty PA, Xiao M, Mills G, Herlyn M, Dong H, Mitchell MJ, Kim J, Xu X, Guo W. Upregulation of exosome secretion from tumor-associated macrophages plays a key role in the suppression of anti-tumor immunity. Cell Rep 2023; 42:113224. [PMID: 37805922 PMCID: PMC10697782 DOI: 10.1016/j.celrep.2023.113224] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 06/15/2023] [Accepted: 09/20/2023] [Indexed: 10/10/2023] Open
Abstract
Macrophages play a pivotal role in tumor immunity. We report that reprogramming of macrophages to tumor-associated macrophages (TAMs) promotes the secretion of exosomes. Mechanistically, increased exosome secretion is driven by MADD, which is phosphorylated by Akt upon TAM induction and activates Rab27a. TAM exosomes carry high levels of programmed death-ligand 1 (PD-L1) and potently suppress the proliferation and function of CD8+ T cells. Analysis of patient melanoma tissues indicates that TAM exosomes contribute significantly to CD8+ T cell suppression. Single-cell RNA sequencing analysis showed that exosome-related genes are highly expressed in macrophages in melanoma; TAM-specific RAB27A expression inversely correlates with CD8+ T cell infiltration. In a murine melanoma model, lipid nanoparticle delivery of small interfering RNAs (siRNAs) targeting macrophage RAB27A led to better T cell activation and sensitized tumors to anti-programmed cell death protein 1 (PD-1) treatment. Our study demonstrates tumors use TAM exosomes to combat CD8 T cells and suggests targeting TAM exosomes as a potential strategy to improve immunotherapies.
Collapse
Affiliation(s)
- Wenqun Zhong
- Department of Biology, School of Arts & Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Youtao Lu
- Department of Biology, School of Arts & Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Xuexiang Han
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jingbo Yang
- Department of Biology, School of Arts & Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Zhiyuan Qin
- Department of Biology, School of Arts & Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Wei Zhang
- Department of Biology, School of Arts & Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ziyan Yu
- Department of Biology, School of Arts & Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Bin Wu
- Department of Biology, School of Arts & Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Shujing Liu
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Wei Xu
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Cathy Zheng
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Lynn M Schuchter
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Giorgos C Karakousis
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Tara C Mitchell
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ravi Amaravadi
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ahron J Flowers
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Phyllis A Gimotty
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Min Xiao
- Molecular and Cellular Oncogenesis Program and Melanoma Research Center, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Gordon Mills
- Division of Oncological Science, School of Medicine, and Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97201, USA
| | - Meenhard Herlyn
- Molecular and Cellular Oncogenesis Program and Melanoma Research Center, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Haidong Dong
- Departments of Urology and Immunology, Mayo College of Medicine and Science, Rochester, MN 55905, USA
| | - Michael J Mitchell
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Junhyong Kim
- Department of Biology, School of Arts & Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Xiaowei Xu
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Wei Guo
- Department of Biology, School of Arts & Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
45
|
Flati I, Di Vito Nolfi M, Dall’Aglio F, Vecchiotti D, Verzella D, Alesse E, Capece D, Zazzeroni F. Molecular Mechanisms Underpinning Immunometabolic Reprogramming: How the Wind Changes during Cancer Progression. Genes (Basel) 2023; 14:1953. [PMID: 37895302 PMCID: PMC10606647 DOI: 10.3390/genes14101953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 10/11/2023] [Accepted: 10/12/2023] [Indexed: 10/29/2023] Open
Abstract
Metabolism and the immunological state are intimately intertwined, as defense responses are bioenergetically expensive. Metabolic homeostasis is a key requirement for the proper function of immune cell subsets, and the perturbation of the immune-metabolic balance is a recurrent event in many human diseases, including cancer, due to nutrient fluctuation, hypoxia and additional metabolic changes occurring in the tumor microenvironment (TME). Although much remains to be understood in the field of immunometabolism, here, we report the current knowledge on both physiological and cancer-associated metabolic profiles of immune cells, and the main molecular circuits involved in their regulation, highlighting similarities and differences, and emphasizing immune metabolic liabilities that could be exploited in cancer therapy to overcome immune resistance.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Daria Capece
- Department of Biotechnological and Applied Clinical Sciences (DISCAB), University of L’Aquila, Via Vetoio, Coppito 2, 67100 L’Aquila, Italy; (I.F.); (M.D.V.N.); (F.D.); (D.V.); (D.V.); (E.A.); (F.Z.)
| | | |
Collapse
|
46
|
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
|
47
|
Li M, Wang M, Wen Y, Zhang H, Zhao G, Gao Q. Signaling pathways in macrophages: molecular mechanisms and therapeutic targets. MedComm (Beijing) 2023; 4:e349. [PMID: 37706196 PMCID: PMC10495745 DOI: 10.1002/mco2.349] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 07/24/2023] [Accepted: 07/27/2023] [Indexed: 09/15/2023] Open
Abstract
Macrophages play diverse roles in development, homeostasis, and immunity. Accordingly, the dysfunction of macrophages is involved in the occurrence and progression of various diseases, such as coronavirus disease 2019 and atherosclerosis. The protective or pathogenic effect that macrophages exert in different conditions largely depends on their functional plasticity, which is regulated via signal transduction such as Janus kinase-signal transducer and activator of transcription, Wnt and Notch pathways, stimulated by environmental cues. Over the past few decades, the molecular mechanisms of signaling pathways in macrophages have been gradually elucidated, providing more alternative therapeutic targets for diseases treatment. Here, we provide an overview of the basic physiology of macrophages and expound the regulatory pathways within them. We also address the crucial role macrophages play in the pathogenesis of diseases, including autoimmune, neurodegenerative, metabolic, infectious diseases, and cancer, with a focus on advances in macrophage-targeted strategies exploring modulation of components and regulators of signaling pathways. Last, we discuss the challenges and possible solutions of macrophage-targeted therapy in clinical applications. We hope that this comprehensive review will provide directions for further research on therapeutic strategies targeting macrophage signaling pathways, which are promising to improve the efficacy of disease treatment.
Collapse
Affiliation(s)
- Ming Li
- Department of Gynecological OncologyTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- National Clinical Research Center for Obstetrics and GynecologyCancer Biology Research Center (Key Laboratory of the Ministry of Education)Tongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Mengjie Wang
- Department of Gynecological OncologyTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- National Clinical Research Center for Obstetrics and GynecologyCancer Biology Research Center (Key Laboratory of the Ministry of Education)Tongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Yuanjia Wen
- Department of Gynecological OncologyTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- National Clinical Research Center for Obstetrics and GynecologyCancer Biology Research Center (Key Laboratory of the Ministry of Education)Tongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Hongfei Zhang
- Department of Gynecological OncologyTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- National Clinical Research Center for Obstetrics and GynecologyCancer Biology Research Center (Key Laboratory of the Ministry of Education)Tongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Guang‐Nian Zhao
- Department of Gynecological OncologyTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- National Clinical Research Center for Obstetrics and GynecologyCancer Biology Research Center (Key Laboratory of the Ministry of Education)Tongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Qinglei Gao
- Department of Gynecological OncologyTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- National Clinical Research Center for Obstetrics and GynecologyCancer Biology Research Center (Key Laboratory of the Ministry of Education)Tongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| |
Collapse
|
48
|
Zeng W, Li F, Jin S, Ho PC, Liu PS, Xie X. Functional polarization of tumor-associated macrophages dictated by metabolic reprogramming. J Exp Clin Cancer Res 2023; 42:245. [PMID: 37740232 PMCID: PMC10517486 DOI: 10.1186/s13046-023-02832-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 09/12/2023] [Indexed: 09/24/2023] Open
Abstract
Macrophages are highly plastic in different tissues and can differentiate into functional subpopulations under different stimuli. Tumor-associated macrophages (TAMs) are one of the most important innate immune cells implicated in the establishment of an immunosuppressive tumor microenvironment (TME). Recent evidence pinpoints the critical role of metabolic reprogramming in dictating pro-tumorigenic functions of TAMs. Both tumor cells and macrophages undergo metabolic reprogramming to meet energy demands in the TME. Understanding the metabolic rewiring in TAMs can shed light on immune escape mechanisms and provide insights into repolarizing TAMs towards anti-tumorigenic function. Here, we discuss how metabolism impinges on the functional divergence of macrophages and its relevance to macrophage polarization in the TME.
Collapse
Affiliation(s)
- Wentao Zeng
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing, 312000, Zhejiang, China
| | - Fei Li
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing, 312000, Zhejiang, China
| | - Shikai Jin
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing, 312000, Zhejiang, China
| | - Ping-Chih Ho
- Department of Fundamental Oncology, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
- Ludwig Lausanne Branch, Lausanne, Switzerland
| | - Pu-Ste Liu
- Institute of Cellular and System Medicine, National Health Research Institute, Miaoli, Taiwan, ROC
| | - Xin Xie
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing, 312000, Zhejiang, China.
| |
Collapse
|
49
|
Brummer C, Pukrop T, Wiskemann J, Bruss C, Ugele I, Renner K. Can Exercise Enhance the Efficacy of Checkpoint Inhibition by Modulating Anti-Tumor Immunity? Cancers (Basel) 2023; 15:4668. [PMID: 37760634 PMCID: PMC10526963 DOI: 10.3390/cancers15184668] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 09/11/2023] [Accepted: 09/17/2023] [Indexed: 09/29/2023] Open
Abstract
Immune checkpoint inhibition (ICI) has revolutionized cancer therapy. However, response to ICI is often limited to selected subsets of patients or not durable. Tumors that are non-responsive to checkpoint inhibition are characterized by low anti-tumoral immune cell infiltration and a highly immunosuppressive tumor microenvironment. Exercise is known to promote immune cell circulation and improve immunosurveillance. Results of recent studies indicate that physical activity can induce mobilization and redistribution of immune cells towards the tumor microenvironment (TME) and therefore enhance anti-tumor immunity. This suggests a favorable impact of exercise on the efficacy of ICI. Our review delivers insight into possible molecular mechanisms of the crosstalk between muscle, tumor, and immune cells. It summarizes current data on exercise-induced effects on anti-tumor immunity and ICI in mice and men. We consider preclinical and clinical study design challenges and discuss the role of cancer type, exercise frequency, intensity, time, and type (FITT) and immune sensitivity as critical factors for exercise-induced impact on cancer immunosurveillance.
Collapse
Affiliation(s)
- Christina Brummer
- Department of Internal Medicine III, Hematology and Oncology, University Hospital Regensburg, 93053 Regensburg, Germany;
- Comprehensive Cancer Center Ostbayern (CCCO), 93053 Regensburg, Germany
| | - Tobias Pukrop
- Department of Internal Medicine III, Hematology and Oncology, University Hospital Regensburg, 93053 Regensburg, Germany;
- Comprehensive Cancer Center Ostbayern (CCCO), 93053 Regensburg, Germany
- Bavarian Cancer Research Center (BZKF), 93053 Regensburg, Germany
| | - Joachim Wiskemann
- National Center for Tumor Diseases (NCT), Heidelberg University Hospital, 69120 Heidelberg, Germany;
| | - Christina Bruss
- Department of Gynecology and Obstetrics, University Medical Center Regensburg, 93053 Regensburg, Germany;
| | - Ines Ugele
- Department of Otorhinolaryngology, University Hospital Regensburg, 93053 Regensburg, Germany; (I.U.); (K.R.)
| | - Kathrin Renner
- Comprehensive Cancer Center Ostbayern (CCCO), 93053 Regensburg, Germany
- Department of Otorhinolaryngology, University Hospital Regensburg, 93053 Regensburg, Germany; (I.U.); (K.R.)
| |
Collapse
|
50
|
Li J, Wang C, Xiao W, Chen Y, Tu J, Wan F, Deng K, Li H. TRAF Family Member 4 Promotes Cardiac Hypertrophy Through the Activation of the AKT Pathway. J Am Heart Assoc 2023; 12:e028185. [PMID: 37642020 PMCID: PMC10547335 DOI: 10.1161/jaha.122.028185] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 05/03/2023] [Indexed: 08/31/2023]
Abstract
Background Pathological cardiac hypertrophy is a major cause of heart failure morbidity. The complex mechanism of intermolecular interactions underlying the pathogenesis of cardiac hypertrophy has led to a lack of development and application of therapeutic methods. Methods and Results Our study provides the first evidence that TRAF4, a member of the tumor necrosis factor receptor-associated factor (TRAF) family, acts as a promoter of cardiac hypertrophy. Here, Western blotting assays demonstrated that TRAF4 is upregulated in cardiac hypertrophy. Additionally, TRAF4 deletion inhibits the development of cardiac hypertrophy in a mouse model after transverse aortic constriction surgery, whereas its overexpression promotes phenylephrine stimulation-induced cardiomyocyte hypertrophy in primary neonatal rat cardiomyocytes. Mechanistically, RNA-seq analysis revealed that TRAF4 promoted the activation of the protein kinase B pathway during cardiac hypertrophy. Moreover, we found that inhibition of protein kinase B phosphorylation rescued the aggravated cardiomyocyte hypertrophic phenotypes caused by TRAF4 overexpression in phenylephrine-treated neonatal rat cardiomyocytes, suggesting that TRAF4 may regulate cardiac hypertrophy in a protein kinase B-dependent manner. Conclusions Our results revealed the regulatory function of TRAF4 in cardiac hypertrophy, which may provide new insights into developing therapeutic and preventive targets for this disease.
Collapse
Affiliation(s)
- Jian Li
- Department of Thoracic and Cardiovascular SurgeryHuanggang Central Hospital of Yangtze UniversityHuanggangChina
| | - Chang‐Quan Wang
- Department of NeurologyHuanggang Central Hospital of Yangtze UniversityHuanggangChina
| | - Wen‐Chang Xiao
- Department of Cardiovascular SurgeryHuanggang Central Hospital of Yangtze UniversityHuanggangChina
- Huanggang Institute of Translational MedicineHuanggangChina
| | - Yun Chen
- Clinical Trial CentersHuanggang Central Hospital of Yangtze UniversityHuanggangChina
| | - Jun Tu
- Huanggang Institute of Translational MedicineHuanggangChina
| | - Feng Wan
- Department of NeurologyHuanggang Central Hospital of Yangtze UniversityHuanggangChina
- Huanggang Institute of Translational MedicineHuanggangChina
| | - Ke‐Qiong Deng
- Huanggang Institute of Translational MedicineHuanggangChina
- Department of CardiologyZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Huo‐Ping Li
- Department of CardiologyHuanggang Central Hospital of Yangtze UniversityHuanggangChina
| |
Collapse
|