1
|
Arya SB, Collie SP, Xu Y, Fernandez M, Sexton JZ, Mosalaganti S, Coulombe PA, Parent CA. Neutrophils secrete exosome-associated DNA to resolve sterile acute inflammation. Nat Cell Biol 2025:10.1038/s41556-025-01671-4. [PMID: 40404894 DOI: 10.1038/s41556-025-01671-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 04/09/2025] [Indexed: 05/24/2025]
Abstract
Acute inflammation, characterized by a rapid influx of neutrophils, is a protective response that can lead to chronic inflammatory diseases when left unresolved. We previously showed that secretion of LTB4-containing exosomes via nuclear envelope-derived multivesicular bodies is required for effective neutrophil infiltration during inflammation. Here we report that the co-secretion of these exosomes with nuclear DNA facilitates the resolution of the neutrophil infiltrate in a mouse skin model of sterile inflammation. Activated neutrophils exhibit rapid and repetitive DNA secretion as they migrate directionally using a mechanism distinct from suicidal neutrophil extracellular trap release and cell death. Packaging of DNA in the lumen of nuclear envelope-multivesicular bodies is mediated by lamin B receptor and chromatin decondensation. These findings advance our understanding of neutrophil functions during inflammation and the physiological relevance of DNA secretion.
Collapse
Affiliation(s)
- Subhash B Arya
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
| | - Samuel P Collie
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
- Cellular and Molecular Biology Graduate Program, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Yang Xu
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Martin Fernandez
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Biophysics, University of Michigan, Ann Arbor, MI, USA
| | - Jonathan Z Sexton
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Medicinal Chemistry, University of Michigan, Ann Arbor, MI, USA
| | - Shyamal Mosalaganti
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Biophysics, University of Michigan, Ann Arbor, MI, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Pierre A Coulombe
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Carole A Parent
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA.
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, USA.
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA.
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, USA.
| |
Collapse
|
2
|
Liu JY, Luo JF, Wu XY, Liu T, Wang R, Zhang Q, Liu YM, Wu H. SLP65/SLP76 Csk-interacting membrane protein promotes hepatic ischemia-reperfusion injury by activating TLR4/Erk1/2-mediated macrophages M1 polarization. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167916. [PMID: 40403937 DOI: 10.1016/j.bbadis.2025.167916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 04/03/2025] [Accepted: 05/19/2025] [Indexed: 05/24/2025]
Abstract
BACKGROUND SLP65/SLP76, Csk-interacting membrane protein (SCIMP), is a membrane protein plays a crucial role in the regulation of macrophage polarization. This study aimed to investigate the mechanism of SCIMP-mediated M1 polarization in liver macrophages and ischemia-reperfusion injury (IRI) in liver transplantation. METHODS Mice underwent orthotopic liver transplantation. In in vivo experiments, mice were divided into the Sham group, LT group, LT+Scramble group, LT+SCIMP (-) group, and LT+ERK1/2 (-) group, and SCIMP or ERK1/2 knockdown was performed using AAV-Erk1/2-RNAi-F4/80-EGFP and AAV-SCIMP-RNAi-F4/80-EGFP. In the subsequent in vitro experiments with primary cells, macrophages were divided into the Ctrl group, H/R group, H/R+Scramble group, H/R+SCIMP (-) group, and H/R+ERK1/2 (-) group, with SCIMP knockdown achieved using siRNA. Immunoprecipitation (IP) was used to detect the interaction between TLR4 and Erk1/2. Liver damage was detected by Hematoxylin and eosin (HE) staining. Polarization was detected by western blot (WB), RT-PCR, immunohistochemistry (IHC), immunofluorescence technique (IF), enzyme-linked immunosorbent assay (ELISA) and flow cytometry (FC). RESULTS Knockdown of SCIMP ameliorated hepatic IRI and liver macrophages M1 polarization. Mechanically, SCIMP promoted the interaction between Erk1/2 and TLR4 in hypoxia/reoxygenation (H/R)-induced liver macrophages, while the inhibition of Erk1/2 reduced liver macrophages M1 polarization and liver IRI. CONCLUSION SCIMP promotes hepatic ischemia-reperfusion injury by activating TLR4/Erk1/2-mediated liver macrophages M1 polarization, which might become a potential therapeutic target in clinic.
Collapse
Affiliation(s)
- Jun-Yan Liu
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, China
| | - Jie-Fu Luo
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, China
| | - Xin-Yi Wu
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, China
| | - Tao Liu
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, China
| | - Rui Wang
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, China
| | - Qi Zhang
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, China
| | - Yi-Ming Liu
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, China.
| | - Hao Wu
- Department of Urology Surgery, The Second Affiliated Hospital of Chongqing Medical University, China.
| |
Collapse
|
3
|
Chen X, Wu C, Tang F, Zhou J, Mo L, Li Y, He J. The Immune Microenvironment: New Therapeutic Implications in Organ Fibrosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e05067. [PMID: 40391706 DOI: 10.1002/advs.202505067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2025] [Revised: 04/28/2025] [Indexed: 05/22/2025]
Abstract
Fibrosis, characterized by abnormal deposition of structural proteins, is a major cause of tissue dysfunction in chronic diseases. The disease burden associated with progressive fibrosis is substantial, and currently approved drugs are unable to effectively reverse it. Immune cells are increasingly recognized as crucial regulators in the pathological process of fibrosis by releasing effector molecules, such as cytokines, chemokines, extracellular vesicles, metabolites, proteases, or intercellular contact. Therefore, targeting the immune microenvironment can be a potential strategy for fibrosis reduction and reversion. This review summarizes the recent advances in the understanding of the immune microenvironment in fibrosis including phenotypic and functional transformations of immune cells and the interaction of immune cells with other cells. The novel opportunities for the discovery and development of drugs for immune microenvironment remodeling and their associated challenges are also discussed.
Collapse
Affiliation(s)
- Xiangqi Chen
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Chuan Wu
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Fei Tang
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jingyue Zhou
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Li Mo
- Center of Gerontology and Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yanping Li
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jinhan He
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
4
|
Aktay-Cetin Ö, Pullamsetti SS, Herold S, Savai R. Lung tumor immunity: redirecting macrophages through infection-induced inflammation. Trends Immunol 2025:S1471-4906(25)00096-1. [PMID: 40382244 DOI: 10.1016/j.it.2025.04.004] [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: 01/05/2025] [Revised: 04/16/2025] [Accepted: 04/18/2025] [Indexed: 05/20/2025]
Abstract
Macrophages play a central role in maintaining tissue homeostasis and in surveillance against pathogens and disease. In the lung, they can adopt either proinflammatory or anti-inflammatory states depending on the nature of the stimulus. As the predominant immune cells in both the lung tumor microenvironment and in sites of lung infection, the functional plasticity of macrophages makes them key players in determining disease outcome. Accurately defining their inflammatory profiles offers an opportunity to reprogram infection-associated macrophages towards enhanced tumor-killing phenotypes. This review explores how acute inflammation can drive macrophage-mediated antitumor immunity and highlights key molecules and signaling pathways that may be leveraged to therapeutically modulate macrophage function.
Collapse
Affiliation(s)
- Öznur Aktay-Cetin
- Institute for Lung Health (ILH), Justus Liebig University, Giessen, Germany; Max Planck Institute for Heart and Lung Research, German Center for Lung Research (DZL), Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany
| | - Soni Savai Pullamsetti
- Institute for Lung Health (ILH), Justus Liebig University, Giessen, Germany; Max Planck Institute for Heart and Lung Research, German Center for Lung Research (DZL), Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany; Department of Internal Medicine II, German Center for Lung Research (DZL), Cardio-Pulmonary Institute (CPI), Justus Liebig University, Giessen, Germany
| | - Susanne Herold
- Institute for Lung Health (ILH), Justus Liebig University, Giessen, Germany; Department of Internal Medicine V, German Center for Lung Research (DZL), German Center for Infection Research (DZIF), Cardio-Pulmonary Institute (CPI), Justus Liebig University, Giessen, Germany
| | - Rajkumar Savai
- Institute for Lung Health (ILH), Justus Liebig University, Giessen, Germany; Max Planck Institute for Heart and Lung Research, German Center for Lung Research (DZL), Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany; Department of Internal Medicine II, German Center for Lung Research (DZL), Cardio-Pulmonary Institute (CPI), Justus Liebig University, Giessen, Germany.
| |
Collapse
|
5
|
Philippon M, Labib R, Ley MBRG, Kaplan LD, Mendez AJ, Best TM, Kouroupis D. Characterization of Extracellular Vesicles from Infrapatellar Fat Pad Mesenchymal Stem/Stromal Cells Expanded Using Regulatory-Compliant Media and Inflammatory/Hormonal Priming. Cells 2025; 14:706. [PMID: 40422209 DOI: 10.3390/cells14100706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2025] [Revised: 04/23/2025] [Accepted: 05/08/2025] [Indexed: 05/28/2025] Open
Abstract
Osteoarthritis (OA) remains a leading cause of disability worldwide, with no disease-modifying therapies currently available for treatment. The infrapatellar fat pad (IFP) harbors mesenchymal stem/stromal cells (MSC) with potent immunomodulatory and regenerative properties, making them a promising candidate for OA treatment. A growing body of evidence suggests that the therapeutic effects of MSC are largely mediated by their extracellular vesicles (EVs), which carry bioactive cargo that modulates inflammation and tissue repair. However, optimizing MSC-derived EVs as a cell-free therapeutic approach requires an in-depth understanding of how culture conditions and inflammatory/hormonal priming influence their functional properties. In this study, IFP-MSC were expanded in regulatory-compliant human platelet lysate (HPL) and xeno-/serum-free (XFSF) media and primed with an inflammatory/fibrotic cocktail (TIC) with oxytocin (OXT) to assess the impact on their immunophenotypic profile and EV cargo. The immunophenotype confirmed that TIC+OXT-primed MSC retained key immunomodulatory surface markers, while EV characterization verified the successful isolation of CD63+/CD9+ vesicles. Pathway enrichment analysis of both HPL- and XFSF- TIC+OXT EVs cargo identified key miRNAs associated with immune regulation, tissue repair, and anabolic signaling. Functional assays revealed that TIC+OXT EVs promoted M2-like anti-inflammatory macrophage polarization and exhibited chondroprotective properties in chondrocytes/synoviocytes inflammatory osteoarthritic assay. These findings highlight the therapeutic potential of TIC+OXT-primed IFP-MSC-derived EVs as immunomodulatory and chondroprotective agents, offering a promising strategy for OA treatment through a clinically viable, cell-free approach.
Collapse
Affiliation(s)
- Marc Philippon
- Department of Orthopaedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, FL 33146, USA
- Diabetes Research Institute & Cell Transplant Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Ramy Labib
- Department of Orthopaedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, FL 33146, USA
- Diabetes Research Institute & Cell Transplant Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Michelle Bellas Romariz Gaudie Ley
- Department of Orthopaedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, FL 33146, USA
- Diabetes Research Institute & Cell Transplant Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- Department of Biomedical Engineering, University of Miami, Miami, FL 33146, USA
| | - Lee D Kaplan
- Department of Orthopaedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, FL 33146, USA
| | - Armando J Mendez
- Diabetes Research Institute & Cell Transplant Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Thomas M Best
- Department of Orthopaedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, FL 33146, USA
| | - Dimitrios Kouroupis
- Department of Orthopaedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, FL 33146, USA
- Diabetes Research Institute & Cell Transplant Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| |
Collapse
|
6
|
Tuersong T, Yong YX, Chen Y, Li PS, Shataer S, Shataer M, Ma LY, Yang XL. Integrating plasma circulating protein-centered multi-omics to identify potential therapeutic targets for Parkinsonian cognitive disorders. J Transl Med 2025; 23:535. [PMID: 40355913 PMCID: PMC12070786 DOI: 10.1186/s12967-025-06541-z] [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: 01/26/2025] [Accepted: 04/25/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND Parkinson's disease (PD), the second most common neurodegenerative disease with notable clinical heterogeneity, has Parkinson disease dementia (PDD) that severely impacts patients' quality of life. As no effective treatment exists, this study aimed to find potential drug targets for PD cognitive disorders. METHODS Two-sample Mendelian randomization (MR) and transcriptome analysis were used to identify PD biomarkers. Protein-protein interaction (PPI), gene ontology (GO), and KEGG pathway analyses explored biological effects. A nomogram model was developed. RESULTS 76 Mendelian randomization genes (MRGs) from MR and 1771 differentially expressed genes (DEGs) from the transcriptome were obtained. Three significant shared DEGs (S-DEGs) were identified, with USP8 and STXBP6 having strong diagnostic value for PDD. The nomogram model with these two genes showed enhanced predictive ability. These genes had physical interactions, co-localization, and correlated with ODC and NEU immune cells. USP8 was linked to five diseases, and STXBP6 to one. CONCLUSION USP8, STXBP6, and immune cells (ODC and NEU) associated with PDD were identified, offering new insights into PD progression.
Collapse
Affiliation(s)
- Tayier Tuersong
- Department of Pharmacy, Xinjiang Key Laboratory of Neurological Diseases, Xinjiang Clinical Research Center for Nervous System Diseases, Second Affiliated Hospital of Xinjiang Medical University, Ürümqi, 830001, Xinjiang, People's Republic of China
| | - Yu Xuan Yong
- Department of Neurology, Xinjiang Clinical Research Center for Nervous System Diseases, Xinjiang Key Laboratory of Neurological Diseases, Second Affiliated Hospital of Xinjiang Medical University, Ürümqi, 830001, Xinjiang, People's Republic of China
| | - Yan Chen
- Department of Pharmacy, Xinjiang Key Laboratory of Neurological Diseases, Xinjiang Clinical Research Center for Nervous System Diseases, Second Affiliated Hospital of Xinjiang Medical University, Ürümqi, 830001, Xinjiang, People's Republic of China
| | - Pei Shan Li
- Department of Neurology, Xinjiang Clinical Research Center for Nervous System Diseases, Xinjiang Key Laboratory of Neurological Diseases, Second Affiliated Hospital of Xinjiang Medical University, Ürümqi, 830001, Xinjiang, People's Republic of China
| | - Samire Shataer
- Department of Neurology, Xinjiang Clinical Research Center for Nervous System Diseases, Xinjiang Key Laboratory of Neurological Diseases, Second Affiliated Hospital of Xinjiang Medical University, Ürümqi, 830001, Xinjiang, People's Republic of China
| | - Munire Shataer
- Department of Histology and Embryology, Basic Medical College of Xinjiang Medical University, Ürümqi, 830001, Xinjiang, People's Republic of China
| | - Liang Ying Ma
- Department of Pharmacy, Xinjiang Key Laboratory of Neurological Diseases, Xinjiang Clinical Research Center for Nervous System Diseases, Second Affiliated Hospital of Xinjiang Medical University, Ürümqi, 830001, Xinjiang, People's Republic of China
| | - Xin Ling Yang
- Department of Neurology, Xinjiang Clinical Research Center for Nervous System Diseases, Xinjiang Key Laboratory of Neurological Diseases, Second Affiliated Hospital of Xinjiang Medical University, Ürümqi, 830001, Xinjiang, People's Republic of China.
| |
Collapse
|
7
|
Xiong B, Wang H, Song YX, Lan WY, Li J, Wang F. Natural saponins and macrophage polarization: Mechanistic insights and therapeutic perspectives in disease management. Front Pharmacol 2025; 16:1584035. [PMID: 40417220 PMCID: PMC12098594 DOI: 10.3389/fphar.2025.1584035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Accepted: 03/24/2025] [Indexed: 05/27/2025] Open
Abstract
Macrophage polarization plays a pivotal role in immune homeostasis and disease progression across inflammatory, neoplastic, and metabolic disorders. Saponins, which are natural compounds with steroidal/triterpenoid structures, demonstrate therapeutic potential through immunomodulatory, anti-inflammatory, and anti-tumor activities. This study aims to highlight the potential of key saponins-such as ginsenosides, astragaloside IV, dioscin, platycodin D, pulsatilla saponins, and panax notoginseng saponins-in modulating macrophage polarization and enhancing conventional therapies, particularly in oncology. We conducted structured searches in PubMed, Google Scholar, and SciFinder (2013-2024) using controlled vocabulary, including "saponins," "macrophage polarization," and "therapeutic effects." Our findings demonstrate that saponins significantly modulate immune responses and improve treatment efficacy. However, clinical translation is hindered by challenges such as poor bioavailability and safety concerns, which limit systemic exposure and therapeutic utility. To overcome these barriers, innovative delivery strategies, including nanoemulsions and engineered exosomes, are essential for enhancing pharmacokinetics and therapeutic index. Future research should prioritize elucidating the molecular mechanisms underlying saponin-mediated macrophage polarization, identifying novel therapeutic targets, and optimizing drug formulations. Addressing these challenges will enable the restoration of immune balance and more effective management of diverse diseases.
Collapse
Affiliation(s)
- Beibei Xiong
- Department of Oncology, The First People’s Hospital of Shuangliu District, Chengdu, China
| | - Huan Wang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yi-Xuan Song
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wen-Ying Lan
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | | | - Fang Wang
- Chengdu First People’s Hospital, Chengdu, China
| |
Collapse
|
8
|
Zhang H, Li M, Zhao B, Chang R, Wang J, Yang Y, Huang Q, Aernouts B, Jiang Q, Loor JJ, Xu C. Evidence for Imbalanced Polarization of Caruncle Macrophages in Retained Placenta of Dairy Cows. J Dairy Sci 2025:S0022-0302(25)00317-0. [PMID: 40349759 DOI: 10.3168/jds.2024-26144] [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: 12/11/2024] [Accepted: 04/11/2025] [Indexed: 05/14/2025]
Abstract
Retained placenta (RP) is a common reproductive disorder with complex etiology and pathogenesis, affecting approximately 8% of dairy cows during the periparturient period. Macrophages constitute 20-25% of all leukocytes at the maternal-fetal interface and coordinate several processes critical for fetal membrane expulsion, including tissue remodeling, induction of apoptosis in damaged cells, and immune activation. This study aimed to investigate the morphological changes at the maternal-fetal interface, as well as the quantity, distribution, and polarization of caruncle macrophages in cows with and without RP. Furthermore, we discuss the potential association between macrophage alterations and histopathological changes in placental tissue of RP cows. A total of 80 Holstein dairy cows (parity, 2-4) were enrolled in this study. Blood samples were collected at -7 d before the expected calving date (-7D), at calving (0h), at 12h postpartum (12h) and at 7 d postpartum (7D). Placental tissue samples were collected within 30 min after parturition. Based on whether the placental membranes were expelled within 12 h postpartum, cows were classified retrospectively into normal expulsion (NE) (n = 6) and RP (n = 6) groups. Picrosirius red staining, along with elevated mRNA and protein levels of Collagen III, indicated enhanced collagen fiber deposition in caruncle tissue. In addition, the mRNA expression of matrix metalloproteinases (MMP-2 and MMP-9) was downregulated in RP tissues, while TIMP-1 was upregulated. Compared with normal expulsion cows, the apoptosis index, as well as the protein and mRNA levels of pro-apoptotic factors (BAX, Caspase-3, Caspase-8) were lower in cows with RP, and the anti-apoptotic factor (BCL2) was higher, indicating reduced apoptosis in the caruncle tissue from RP cows. In both the serum and tissues, we observed lower levels of chemotactic factors (CXCL1 and MCP-1) in RP cows, alongside increased IL-10 (an immunosuppressive factor) and decreased IL-1β (an immune-stimulatory factor). The downregulated protein and mRNA abundance of the macrophage marker CD68, consistent with reduced presence of CD68+ cells observed through immunofluorescence, revealed low numbers of caruncle macrophages in cows with RP. Further, the caruncles tissue of RP cows displayed significant alterations in the distribution of CD68+ macrophages, with reduced infiltration into trophoblast cells. Regarding macrophage phenotypic changes in RP cows, the greater protein and mRNA expression of M2 polarization markers (CD206, IL-10, IL-6, and TGF-β) along with greater numbers of CD206+/CD68+ cells detected through immunofluorescence indicated that macrophage polarization phenotype in the caruncles of RP cows shifted predominantly toward M2 phenotype. In contrast, RP cows exhibited lower protein and mRNA levels of M1 polarization markers (CD86, iNOS, IL-1β, and NF-κB), as well as reduced numbers of CD86+/CD68+ cells. Overall, caruncle tissues from RP cows were characterized by a reduced macrophage population with a predominant M2 phenotype. Alterations in the quantity and polarization state of macrophages at the maternal-fetal interface may lead to reduced immune cell trafficking into the caruncle, thus impairing the apoptotic and proteolytic processes essential for placental expulsion.
Collapse
Affiliation(s)
- Huijing Zhang
- College of Veterinary Medicine, China Agricultural University, 100193, Beijing, China
| | - Ming Li
- College of Veterinary Medicine, China Agricultural University, 100193, Beijing, China
| | - Bichen Zhao
- College of Veterinary Medicine, China Agricultural University, 100193, Beijing, China
| | - Renxu Chang
- College of Veterinary Medicine, China Agricultural University, 100193, Beijing, China
| | - Jingyi Wang
- College of Veterinary Medicine, China Agricultural University, 100193, Beijing, China
| | - Yue Yang
- College of Veterinary Medicine, China Agricultural University, 100193, Beijing, China
| | - Qingnian Huang
- College of Veterinary Medicine, China Agricultural University, 100193, Beijing, China
| | - Ben Aernouts
- Department of Biosystems, Division of Animal and Human Health Engineering, KU Leuven, Geel Campus, Kleinhoefstraat 4, 2440 Geel, Belgium
| | - Qianming Jiang
- Mammalian NutriPhysio Genomics, Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana 61801
| | - Juan J Loor
- Mammalian NutriPhysio Genomics, Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana 61801
| | - Chuang Xu
- College of Veterinary Medicine, China Agricultural University, 100193, Beijing, China..
| |
Collapse
|
9
|
Karagiannis GS, Kato Y, Entenberg D. Editorial: Exploring macrophage roles in cancer progression and therapeutic targeting. Front Immunol 2025; 16:1612588. [PMID: 40406110 PMCID: PMC12095945 DOI: 10.3389/fimmu.2025.1612588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2025] [Accepted: 04/25/2025] [Indexed: 05/26/2025] Open
Affiliation(s)
- George S. Karagiannis
- Integrated Imaging Program for Cancer Research, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, United States
- Cancer Dormancy Institute, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, United States
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, United States
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, United States
- Tumor Microenvironment and Metastasis Program, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, United States
| | - Yu Kato
- Tsukuba Research Laboratories, Eisai Co., Ltd., Tsukuba, Ibaraki, Japan
| | - David Entenberg
- Integrated Imaging Program for Cancer Research, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, United States
- Cancer Dormancy Institute, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, United States
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, United States
- Tumor Microenvironment and Metastasis Program, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, United States
- Department of Pathology, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, United States
| |
Collapse
|
10
|
Tao M, Wang L, Chen C, Tang M, Wang Y, Zhang J, Zhao X, Feng Q, Chen J, Yan W, Lin R, Fu Y. Developmentally endothelial locus-1 facilitates intestinal inflammation resolution by suppressing the Cmpk2-cGAS-STING pathway and promoting reparatory macrophage transition. J Adv Res 2025:S2090-1232(25)00274-7. [PMID: 40288675 DOI: 10.1016/j.jare.2025.04.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 03/25/2025] [Accepted: 04/18/2025] [Indexed: 04/29/2025] Open
Abstract
INTRODUCTION Abnormalities in inflammation resolution function are intimately linked to chronic inflammation, and proresolution therapies may offer novel opportunities for IBD treatment. Developmental endothelial locus 1 (DEL-1), a natural modulator of tissue immunity and inflammation resolution, has not been studied in IBD. OBJECTIVES We aimed to investigate the expression and functions of DEL-1 in IBD. METHODS Assessment of DEL-1 expression in patients, murine models, and cellular levels. To explore the effects of DEL-1 in the acute and recovery phases of inflammation, overexpression plasmids, adeno-associated viruses for DEL-1 knockdown, and DEL-1-Fc fusion proteins were administered to cells and mice. Additionally, the potential mechanism of DEL-1 in IBD was demonstrated using flow cytometry, RNA-Seq, ChIP, dual-luciferase reporter assays and 16S rRNA. RESULTS DEL-1 levels were significantly reduced in IBD patients, colitis mice and macrophages, while the levels increased with inflammation to resolve. Transfection with DEL-1 overexpression plasmid or DEL-1-Fc intervention reduces levels of inflammatory cytokines in both phases and upregulates reparative gene levels in the recovery phase. DEL-1 knockdown inhibits inflammation resolution of colitis. Mechanistically, we demonstrated that DEL-1 inhibits Cmpk2-dependent mtDNA synthesis, thereby inhibiting the cGAS-STING pathway to ameliorate intestinal inflammation. Moreover, DEL-1 promotes reparative macrophage transition in the repair model of colitis. Spi1 was identified as a transcription factor that regulates Cmpk2 and the reparative gene Il10. Intervention with overexpression plasmid of Spi1 or Cmpk2 or the STING agonist DMXAA reverses the effects of DEL-1. In parallel, DEL-1 also inhibits neutrophil recruitment, repairs the intestinal barrier, and improves intestinal microbiota dysbiosis. CONCLUSION We report the first demonstration that DEL-1 significantly ameliorates colonic inflammation in colitis mice. Our findings elucidate a novel mechanism wherein DEL-1 exerts its protective effects by suppressing the Cmpk2-cGAS-STING pathway and promoting reparative macrophage transition. These results collectively position DEL-1 as a promising therapeutic avenue for IBD.
Collapse
Affiliation(s)
- Meihui Tao
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li Wang
- Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chaoyue Chen
- Department of Gastroenterology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Mengfan Tang
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yanping Wang
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jingyue Zhang
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xi Zhao
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qinyu Feng
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Junfa Chen
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Yan
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Rong Lin
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Yu Fu
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
11
|
Jeong MR, Hwang JW, Choi M, Seok SH. MHC class II + macrophage differentiation is impaired in metastasized lungs via PGE 2 receptor EP2. Cell Rep 2025; 44:115574. [PMID: 40232933 DOI: 10.1016/j.celrep.2025.115574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 01/17/2025] [Accepted: 03/26/2025] [Indexed: 04/17/2025] Open
Abstract
Monocytes differentiate into macrophages (Mφs) to facilitate lung metastasis, but the monocyte-to-Mφ transition during this process is not well understood. To investigate, we performed bulk RNA sequencing on Mφs isolated from the lungs of mice bearing Lewis lung carcinoma tumors and from naive lungs. Our results showed impaired differentiation of monocytes into major histocompatibility complex (MHC) class II+ Mφs, with an upregulation of PGE2-inducible genes, including Arg1, in tumor-associated Mφs (TAMs). In vitro experiments confirmed that prostaglandin E2 (PGE2) inhibits the differentiation of MHC class II+ Mφs while promoting Arg1+ Mφs via the E prostanoid 2 (EP2) receptor, accompanied by DNA methylation. Whole-genome bisulfite sequencing revealed that PGE2-EP2 signaling drives the hypermethylation and downregulation of gene sets related to myeloid cells in non-neoplastic tissues. Our study highlights PGE2-EP2-driven DNA methylation in the monocyte-to-TAM transition, suggesting potential therapeutic avenues for lung metastasis.
Collapse
Affiliation(s)
- Mi Reu Jeong
- Macrophage Lab, Department of Microbiology and Immunology, and Institute of Endemic Disease, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Jung Woo Hwang
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Murim Choi
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Seung Hyeok Seok
- Macrophage Lab, Department of Microbiology and Immunology, and Institute of Endemic Disease, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; Cancer Research Institute, Seoul National University, Seoul 03080, Republic of Korea.
| |
Collapse
|
12
|
Fu X, Zhang L, Lin J, Wang Q, Wang Z, Chi M, Li D, Zhao G, Li C. Zeolitic Imidazolate Framework-90 Treats Fungal Keratitis by Promoting Macrophage Apoptosis and Targeting Increased Mitochondrial Reactive Oxygen Species in Aspergillus Fumigatus. Int J Nanomedicine 2025; 20:4551-4569. [PMID: 40242606 PMCID: PMC12002346 DOI: 10.2147/ijn.s517169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Accepted: 04/07/2025] [Indexed: 04/18/2025] Open
Abstract
Background Fungal keratitis is a severe vision-threatening corneal infection with a prognosis influenced by fungal virulence and the host's immune defense mechanisms. However, there is still a lack of effective drugs that attenuate fungal virulence while relieving the inflammatory response caused by fungal keratitis. Purpose Finding an effective treatment to solve these problems is particularly important. Methods We synthesized Zeolitic imidazolate framework-90 (ZIF-90) by water-based synthesis method and characterized it. In vitro experiments included mycelium electron microscopy, Cell Counting Kit-8 (CCK-8), and Enzyme-linked immunosorbent assay (ELISA). These trials verified the disruptive effects of ZIF-90 on morphology, cell membrane, cell wall, and biofilm formation of Aspergillus fumigatus (A. fumigatus). These experiments also demonstrated the impact of ZIF-90 on the proinflammatory cytokines tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6). Moreover, the effect of ZIF-90 on mitochondrial reactive oxygen species (mtROS) of cells and fungi was verified by MitoSOX Red Mitochondrial Superoxide Indicator (MitoSOX). In vivo, corneal toxicity test, establishment and treatment of mycotic keratitis mouse model, and immunofluorescence staining were used to evaluate the efficacy of ZIF-90 in the procedure of fungal keratitis. In addition, to investigate whether the metal-ligand zinc and the organic ligand imidazole acted as essential factors in ZIF-90, we investigated the in vitro antimicrobial and anti-inflammatory effects of ZIF-8, ZIF-67 and Metal-Organic Frameworks-74 (Zn) (MOF-74 (Zn)) by Minimum Inhibitory Concentration (MIC) and ELISA experiments. Results ZIF-90 has therapeutic effects on fungal keratitis, which could break the protective organelles of A. fumigatus, such as the cell wall. In addition, ZIF-90 can also be targeted to increase the amount of mtROS in fungi and promote apoptosis of macrophages. The results demonstrated that both zinc ions and imidazole possessed antimicrobial and anti-inflammatory effects. In addition, ZIF-90 exhibited better antifungal properties than ZIF-8, ZIF-67, and MOF-74 (Zn).
Collapse
Affiliation(s)
- Xueyun Fu
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
| | - Lina Zhang
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
| | - Jing Lin
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
| | - Qian Wang
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
| | - Ziyi Wang
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
| | - Menghui Chi
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
| | - Daohao Li
- State Key Laboratory of Bio-fibers and Eco-Textiles, Institute of Marine Biobased Materials, College of Materials Science and Engineering, Qingdao University, Qingdao, People’s Republic of China
| | - Guiqiu Zhao
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
| | - Cui Li
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
| |
Collapse
|
13
|
Tan Y, Yang YG, Zhang X, Zhao L, Wang X, Liu W. Tumor cell-derived osteopontin promotes tumor fibrosis indirectly via tumor-associated macrophages. J Transl Med 2025; 23:432. [PMID: 40217301 PMCID: PMC11992893 DOI: 10.1186/s12967-025-06444-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Accepted: 03/28/2025] [Indexed: 04/14/2025] Open
Abstract
BACKGROUND High fibrosis of the tumor microenvironment (TME) not only impedes the effective infiltration of T cells but also serves as a physical barrier to inhibit the penetration of chemotherapy drugs. Triple-negative breast cancer (TNBC) is characterized by significant infiltration of tumor-associated macrophages (TAMs) and high fibrosis. However, the mechanism of high fibrosis in such tumors is still under debate. METHODS We first investigated the correlation between tumor-derived osteopontin (OPN) and tumor fibrosis as well as TAM enrichment using a tumor model characterized by OPN genetic inactivation or overexpression. We further compared the effects of macrophage depletion on tumor fibrosis in mice bearing TNBC tumors (4T1WT or 4T1Spp1 - KO). To elucidate the mechanism by which TAMs promote tumor fibrosis, we evaluated their potential to recruit cancer-associated fibroblasts (CAFs) through in vitro migration assays and compared the production of transforming growth factor-beta 1 (TGFβ1) among different TAM subpopulations. RESULTS Our study revealed that OPN secretion by tumor cells correlates positively with both tumor fibrosis and TAM enrichment. Specifically, within the enriched TAM population, Ly6C+CD206- TAMs recruit CAFs via CCL5 secretion, while Ly6C-CD206high TAMs secrete TGFβ1 to activate CAFs. Blocking the tumor cell-derived OPN can effectively prevent tumor fibrosis. CONCLUSIONS This study shows that tumor-derived OPN primarily drives TAM enrichment in mouse cancer model, indirectly promoting tumor fibrosis through Ly6C+CD206-/low and Ly6C-CD206high TAMs. Our findings have potential application in preventing tumors from excessive fibrosis and enhancing the efficacy of immunotherapy and chemotherapy.
Collapse
Affiliation(s)
- Yuying Tan
- Key Laboratory of Organ Regeneration & Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Jilin University, Changchun, China
- Echocardiography Department, The First Hospital of Jilin University, Changchun, China
| | - Yong-Guang Yang
- Key Laboratory of Organ Regeneration & Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Jilin University, Changchun, China
- International Center of Future Science, Jilin University, Changchun, China
| | - Xiaoying Zhang
- Key Laboratory of Organ Regeneration & Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Jilin University, Changchun, China
| | - Lei Zhao
- Key Laboratory of Organ Regeneration & Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Jilin University, Changchun, China
| | - Xiaocong Wang
- Echocardiography Department, The First Hospital of Jilin University, Changchun, China.
| | - Wentao Liu
- Key Laboratory of Organ Regeneration & Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, China.
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Jilin University, Changchun, China.
| |
Collapse
|
14
|
Posey JN, Jordan M, Lewis CV, Sul C, Dobrinskikh E, Swindle D, Denorme F, Irwin D, Di Paola J, Stenmark K, Nozik ES, Delaney C. Nbeal2 knockout mice are not protected against hypoxia-induced pulmonary vascular remodeling and pulmonary hypertension. Blood Adv 2025; 9:1571-1584. [PMID: 39693512 PMCID: PMC11986223 DOI: 10.1182/bloodadvances.2024013880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 11/05/2024] [Accepted: 11/29/2024] [Indexed: 12/20/2024] Open
Abstract
ABSTRACT Inflammation drives the initiation and progression of pulmonary hypertension (PH). Platelets, increasingly recognized as immune cells, are activated and increased in the lungs of patients with PH. Platelet activation leads to the release of α-granule chemokines, many of which are implicated in PH. We hypothesized that hypoxia-induced secretion of platelet α-granule-stored proteins and PH would be prevented in Neurobeachin-like 2 knockout (Nbeal2-/-) α-granule-deficient mice. Wild-type (WT) and Nbeal2-/- mice were maintained in normoxia or exposed to 10% hypobaric hypoxia for 3, 14, 21, or 35 days. We observed macrothrombocytopenia, increased circulating neutrophils and monocytes, and increased lung interstitial macrophages (IMs) in Nbeal2-/- mice at baseline. Hypoxia-induced platelet activation was attenuated, and hypoxia-induced increase in lung platelet factor 4 (PF4) and platelets was delayed in Nbeal2-/- mice compared with in WT mice. Finally, although pulmonary vascular remodeling (PVR) and PH were attenuated at day 21, Nbeal2-/- mice were not protected against hypoxia-induced PVR and PH at day 35. Although this mutation also affected circulating monocytes, neutrophils, and lung IMs, all of which are critical in the development of experimental PH, we gained further support for the role of platelets and α-granule proteins, such as PF4, in PH progression and pathogenesis and made several observations that expand our understanding of α-granule-deficient mice in chronic hypoxia.
Collapse
Affiliation(s)
- Janelle N. Posey
- Cardiovascular Pulmonary Research Laboratory, University of Colorado Anschutz Medical Campus, Aurora, CO
- Section of Neonatology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Mariah Jordan
- Cardiovascular Pulmonary Research Laboratory, University of Colorado Anschutz Medical Campus, Aurora, CO
- Section of Neonatology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Caitlin V. Lewis
- Cardiovascular Pulmonary Research Laboratory, University of Colorado Anschutz Medical Campus, Aurora, CO
- Section of Pediatric Critical Care, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Christina Sul
- Cardiovascular Pulmonary Research Laboratory, University of Colorado Anschutz Medical Campus, Aurora, CO
- Section of Pediatric Critical Care, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Evgenia Dobrinskikh
- Cardiovascular Pulmonary Research Laboratory, University of Colorado Anschutz Medical Campus, Aurora, CO
- Section of Neonatology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Delaney Swindle
- Cardiovascular Pulmonary Research Laboratory, University of Colorado Anschutz Medical Campus, Aurora, CO
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Frederik Denorme
- Department of Emergency Medicine, Washington University School of Medicine, St. Louis, MO
| | - David Irwin
- Cardiovascular Pulmonary Research Laboratory, University of Colorado Anschutz Medical Campus, Aurora, CO
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Jorge Di Paola
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, Washington University in St. Louis, St. Louis, MO
| | - Kurt Stenmark
- Cardiovascular Pulmonary Research Laboratory, University of Colorado Anschutz Medical Campus, Aurora, CO
- Section of Pediatric Critical Care, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Eva S. Nozik
- Cardiovascular Pulmonary Research Laboratory, University of Colorado Anschutz Medical Campus, Aurora, CO
- Section of Pediatric Critical Care, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Cassidy Delaney
- Cardiovascular Pulmonary Research Laboratory, University of Colorado Anschutz Medical Campus, Aurora, CO
- Section of Neonatology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO
| |
Collapse
|
15
|
Liu C, Xu L, Feng J, Yang B, Chen K, Liu Y, Wu X, Wu S, Li Z, Chen S, Chen Z. Fluorine-Incorporated Biogenic Hydroxyapatite Enhances Socket Bone Healing via Addressing Macrophage-Mediated Inflammatory Response. Bioengineering (Basel) 2025; 12:396. [PMID: 40281756 PMCID: PMC12024827 DOI: 10.3390/bioengineering12040396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 03/26/2025] [Accepted: 04/01/2025] [Indexed: 04/29/2025] Open
Abstract
Biological hydroxyapatite (BHA) has been extensively employed in alveolar socket preservation, yet its clinical application is often compromised by delayed bone healing triggered by macrophage-mediated pro-inflammatory responses. Building upon our previous work, in which we successfully incorporated fluorine into BHA to develop fluorinated biogenic hydroxyapatite (FBHA) with superior physicochemical and biological properties, this study systematically investigated the effects of fluorine doping on macrophage-mediated osteoimmunomodulation and socket bone healing. The synthesized FBHA was characterized using SEM, EDS, and fluoride ion release assays to confirm fluorine incorporation. In macrophage co-culture models, FBHA demonstrated significant advantages over BHA, effectively suppressing iNOS and TNFα gene expression, reducing NO release, and inhibiting phagocytic activity in M1 macrophages. RNAseq analysis revealed that the M1 phenotype suppression might be mediated through enhanced cellular antioxidant activity. Moreover, in macrophage-conditioned microenvironments, FBHA significantly upregulated osteogenic gene expression and ALP activity of pre-osteoblasts. In vivo experiments demonstrated FBHA's superior performance in alveolar ridge preservation, especially in new bone formation and mineralization inside sockets. Fluorine doping significantly boosted socket bone healing via suppressing the inflammatory response of macrophages and enhancing osteogenic differentiation of pre-osteoblasts. These findings provide valuable insights into the development of next-generation biomaterials for alveolar socket preservation.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Shoucheng Chen
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University and Guangdong Provincial Key Laboratory of Stomatology, Guangdong Research Center for Dental and Cranial Rehabilitation and Material Engineering, Guangzhou 510055, China; (C.L.)
| | - Zhuofan Chen
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University and Guangdong Provincial Key Laboratory of Stomatology, Guangdong Research Center for Dental and Cranial Rehabilitation and Material Engineering, Guangzhou 510055, China; (C.L.)
| |
Collapse
|
16
|
Wachter E, Fox LH, Lu Z, Jones AD, Casto ND, Waltz SE. RON Receptor Signaling and the Tumor Microenvironment. Genes (Basel) 2025; 16:437. [PMID: 40282397 PMCID: PMC12026484 DOI: 10.3390/genes16040437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2025] [Revised: 04/01/2025] [Accepted: 04/03/2025] [Indexed: 04/29/2025] Open
Abstract
The immune microenvironment plays a critical role in tumor growth and development. Immune activation within the tumor microenvironment is dynamic and can be modulated by tumor intrinsic and extrinsic signaling. The RON receptor tyrosine kinase is canonically associated with growth signaling and wound healing, and this receptor is frequently overexpressed in a variety of cancers. Epithelial cells, macrophages, dendritic cells, and fibroblasts express RON, presenting an important axis by which RON overexpressing tumors influence the tumor microenvironment. This review synthesizes the existing literature on the roles of tumor cell-intrinsic and -extrinsic RON signaling, highlighting areas of interest and gaps in knowledge that show potential for future studies.
Collapse
Affiliation(s)
- Emily Wachter
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267-0521, USA
| | - Levi H. Fox
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267-0521, USA
| | - Zhixin Lu
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267-0521, USA
| | - Angelle D. Jones
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267-0521, USA
| | - Nicholas D. Casto
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267-0521, USA
| | - Susan E. Waltz
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267-0521, USA
- Research Service, Cincinnati Veterans Affairs Hospital Medical Center, Cincinnati, OH 45220, USA
| |
Collapse
|
17
|
Lohia GK, Riquelme SA. Influence of cell bioenergetics on host-pathogen interaction in the lung. Front Immunol 2025; 16:1549293. [PMID: 40248701 PMCID: PMC12003392 DOI: 10.3389/fimmu.2025.1549293] [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: 12/20/2024] [Accepted: 03/19/2025] [Indexed: 04/19/2025] Open
Abstract
Pulmonary diseases, arising from infections caused by bacteria, fungi, and viruses, or stemming from underlying genetic factors are one of the leading causes of mortality in humans, accounting for millions of deaths every year. At the onset of pulmonary diseases, crucial roles are played by phagocytic immune cells, particularly tissue-resident macrophages, in regulating the immune response at the mucosal barrier. Recent strides have illuminated the pivotal role of host bioenergetics modulated by metabolites derived from both pathogens and hosts in influencing the pathophysiology of major organs. Their influence extends to processes such as the infiltration of immune cells, activation of macrophages, and the polarization phenomenon. Furthermore, host-derived metabolites, such as itaconate, contribute to the promotion of anti-inflammatory responses, thereby preventing immunopathology and facilitating the preservation of mucosal niches to thrive for the long-term. This review explores recent advancements in the field of immunometabolism, with a particular emphasis on the intricacies of disease progression in pulmonary infections caused by bacteria such as P. aeruginosa, M. tuberculosis and S. aureus and fungi like C. albicans.
Collapse
|
18
|
Iliakis CS, Crotta S, Wack A. The Interplay Between Innate Immunity and Nonimmune Cells in Lung Damage, Inflammation, and Repair. Annu Rev Immunol 2025; 43:395-422. [PMID: 40036704 DOI: 10.1146/annurev-immunol-082323-031852] [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: 03/06/2025]
Abstract
As the site of gas exchange, the lung is critical for organismal survival. It is also subject to continual environmental insults inflicted by pathogens, particles, and toxins. Sometimes, these insults result in structural damage and the initiation of an innate immune response. Operating in parallel, the immune response aims to eliminate the threat, while the repair process ensures continual physiological function of the lung. The inflammatory response and repair processes are thus inextricably linked in time and space but are often studied in isolation. Here, we review the interplay of innate immune cells and nonimmune cells during lung insult and repair. We highlight how cellular cross talk can fine-tune the circuitry of the immune response, how innate immune cells can facilitate or antagonize proper organ repair, and the prolonged changes to lung immunity and physiology that can result from acute immune responses and repair processes.
Collapse
Affiliation(s)
- Chrysante S Iliakis
- Immunoregulation Laboratory, The Francis Crick Institute, London, United Kingdom;
| | - Stefania Crotta
- Immunoregulation Laboratory, The Francis Crick Institute, London, United Kingdom;
| | - Andreas Wack
- Immunoregulation Laboratory, The Francis Crick Institute, London, United Kingdom;
| |
Collapse
|
19
|
Zou Y, Cao M, Tai M, Zhou H, Tao L, Wu S, Yang K, Zhang Y, Ge Y, Wang H, Luo S, Ju Z. A Feedback Loop Driven by H4K12 Lactylation and HDAC3 in Macrophages Regulates Lactate-Induced Collagen Synthesis in Fibroblasts Via the TGF-β Signaling. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2411408. [PMID: 39945346 PMCID: PMC11967864 DOI: 10.1002/advs.202411408] [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: 09/17/2024] [Revised: 01/20/2025] [Indexed: 04/05/2025]
Abstract
The decrease in fibroblast collagen is a primary contributor to skin aging. Lactate can participate in collagen synthesis through lysine lactylation by regulating gene transcription. However, the precise mechanism by which lactate influences collagen synthesis requires further investigation. This study demonstrates that the depletion of macrophages mitigates the stimulating effect of lactate on collagen synthesis in fibroblasts. Through joint CUT&Tag and RNA-sequencing analyses, a feedback loop between H4K12 lactylation (H4K12la) and histone deacetylase 3 (HDAC3) in macrophages that drives lactate-induced collagen synthesis are identified. Macrophages can uptake extracellular lactate via monocarboxylate transporter-1 (MCT1), leading to an up-regulation of H4K12la levels through a KAT5-KAT8-dependent mechanism in response to Poly-L-Lactic Acid (PLLA) stimulation, a source of low concentration and persistent lactate, thereby promoting collagen synthesis in fibroblasts. Furthermore, H4K12la is enriched at the promoters of TGF-β1 and TGF-β3, enhancing their transcription. Hyperlactylation of H4K12la inhibits the expression of the eraser HDAC3, while the activation of HDAC3 reduces H4K12la in macrophages and suppresses collagen synthesis in fibroblasts. In conclusion, this study illustrates that macrophages play a critical role in lactate-induced collagen synthesis in the skin, and targeting the lactate-H4K12la-HDAC3-TGF-β axis may represent a novel approach for enhancing collagen production to combat skin aging.
Collapse
Affiliation(s)
- Ying Zou
- Key Laboratory of Regenerative Medicine of Ministry of EducationInstitute of Aging and Regenerative MedicineCollege of Life Science and TechnologyJinan UniversityGuangzhou510632China
- Department of Plastic and Reconstructive SurgeryGuangdong Second Provincial General HospitalJinan UniversityGuangzhou510403China
| | - Mibu Cao
- Department of Plastic and Reconstructive SurgeryGuangdong Second Provincial General HospitalJinan UniversityGuangzhou510403China
| | - Meiling Tai
- R&D CenterInfinitus (China) Company LtdGuangzhou510640China
| | - Haoxian Zhou
- Department of CardiologyGuangdong Provincial Cardiovascular InstituteGuangdong Provincial People's HospitalGuangdong Academy of Medical SciencesGuangzhou510080China
| | - Li Tao
- Key Laboratory of Regenerative Medicine of Ministry of EducationInstitute of Aging and Regenerative MedicineCollege of Life Science and TechnologyJinan UniversityGuangzhou510632China
| | - Shu Wu
- Key Laboratory of Regenerative Medicine of Ministry of EducationInstitute of Aging and Regenerative MedicineCollege of Life Science and TechnologyJinan UniversityGuangzhou510632China
| | - Kaiye Yang
- R&D CenterInfinitus (China) Company LtdGuangzhou510640China
| | - Youliang Zhang
- Department of Plastic and Reconstructive SurgeryGuangdong Second Provincial General HospitalJinan UniversityGuangzhou510403China
| | - Yuanlong Ge
- Key Laboratory of Regenerative Medicine of Ministry of EducationInstitute of Aging and Regenerative MedicineCollege of Life Science and TechnologyJinan UniversityGuangzhou510632China
| | - Hao Wang
- Department of AnesthesiologyThe First Affiliated HospitalJinan UniversityGuangzhou510632China
| | - Shengkang Luo
- Key Laboratory of Regenerative Medicine of Ministry of EducationInstitute of Aging and Regenerative MedicineCollege of Life Science and TechnologyJinan UniversityGuangzhou510632China
- Department of Plastic and Reconstructive SurgeryGuangdong Second Provincial General HospitalJinan UniversityGuangzhou510403China
| | - Zhenyu Ju
- Key Laboratory of Regenerative Medicine of Ministry of EducationInstitute of Aging and Regenerative MedicineCollege of Life Science and TechnologyJinan UniversityGuangzhou510632China
- Department of Plastic and Reconstructive SurgeryGuangdong Second Provincial General HospitalJinan UniversityGuangzhou510403China
| |
Collapse
|
20
|
Zhou Y, Wen T, Yang S, Meng B, Wei J, Zhang J, Wang L, Shen X. Sesquiterpene lactones from Cichorium intybus exhibit potent anti-inflammatory and hepatoprotective effects by repression of NF-κB and enhancement of NRF2. JOURNAL OF ETHNOPHARMACOLOGY 2025; 343:119439. [PMID: 39904423 DOI: 10.1016/j.jep.2025.119439] [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: 10/24/2024] [Revised: 01/26/2025] [Accepted: 01/31/2025] [Indexed: 02/06/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Cichorium intybus is a traditional medicinal herb for hepatitis treatment in China and Europe. Sesquiterpene lactones are the main active ingredients in C. intybus. However, their structure-activity relationship (SAR) and molecular mechanisms of anti-inflammatory and hepatoprotective effects require further elucidation. AIM OF THE STUDY To identify new sesquiterpene lactones from C. intybus, and further evaluate their anti-inflammatory effects, SAR, and mechanisms of anti-inflammatory and hepatoprotective properties. METHODS Identification of sesquiterpene lactones from C. intybus using chromatographic fractionation, NMR, and mass spectrometry. The repression of inflammation was evaluated in RAW264.7 macrophages incubated with LPS. Western blotting was employed to investigate the anti-inflammatory mechanisms. The hepatoprotective effect was measured in LPS/D-galactosamine (D-GalN)-induced acute hepatitis in mice. RESULTS We identified 3 new sesquiterpene lactones and 15 known analogues from C. intybus. SAR analysis showed that the α-methylene-γ-lactone moiety was essential for their anti-inflammatory properties. Furthermore, 8-deoxylactucin was identified as the most potent anti-inflammatory component in LPS-induced RAW264.7 macrophages by reduction of nitric oxide production via inhibiting iNOS expression, and suppression of IL-1β, IL-6, and TNF-α expression. Mechanistically, 8-deoxylactucin not only blocked LPS-induced IKKα/β phosphorylation, IκBα phosphorylation and degradation, and NF-κB nuclear accumulation, but also enhanced NRF2 expression and nuclear translocation, HO-1 and NQO1 expression, and reduced ROS generation in vitro. In vivo, 8-deoxylactucin mitigated LPS/D-GalN-induced acute hepatitis, which manifested as reduction in inflammatory infiltration, live injury, serum levels of AST and ALT, and production of pro-inflammatory cytokines and 4-hydroxynonenal. CONCLUSION 8-Deoxylactucin, the sesquiterpene lactone isolated from C. intybus, exerted anti-inflammatory and hepatoprotective effects by blocking NF-κB activation and enhancing NRF2 activation.
Collapse
Affiliation(s)
- Yan Zhou
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Tian Wen
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China; College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Shan Yang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Binru Meng
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jing Wei
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China; College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jing Zhang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Lun Wang
- Center for Natural Products Research, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, China.
| | - Xiaofei Shen
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| |
Collapse
|
21
|
Guan F, Wang R, Yi Z, Luo P, Liu W, Xie Y, Liu Z, Xia Z, Zhang H, Cheng Q. Tissue macrophages: origin, heterogenity, biological functions, diseases and therapeutic targets. Signal Transduct Target Ther 2025; 10:93. [PMID: 40055311 PMCID: PMC11889221 DOI: 10.1038/s41392-025-02124-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 11/01/2024] [Accepted: 12/15/2024] [Indexed: 05/04/2025] Open
Abstract
Macrophages are immune cells belonging to the mononuclear phagocyte system. They play crucial roles in immune defense, surveillance, and homeostasis. This review systematically discusses the types of hematopoietic progenitors that give rise to macrophages, including primitive hematopoietic progenitors, erythro-myeloid progenitors, and hematopoietic stem cells. These progenitors have distinct genetic backgrounds and developmental processes. Accordingly, macrophages exhibit complex and diverse functions in the body, including phagocytosis and clearance of cellular debris, antigen presentation, and immune response, regulation of inflammation and cytokine production, tissue remodeling and repair, and multi-level regulatory signaling pathways/crosstalk involved in homeostasis and physiology. Besides, tumor-associated macrophages are a key component of the TME, exhibiting both anti-tumor and pro-tumor properties. Furthermore, the functional status of macrophages is closely linked to the development of various diseases, including cancer, autoimmune disorders, cardiovascular disease, neurodegenerative diseases, metabolic conditions, and trauma. Targeting macrophages has emerged as a promising therapeutic strategy in these contexts. Clinical trials of macrophage-based targeted drugs, macrophage-based immunotherapies, and nanoparticle-based therapy were comprehensively summarized. Potential challenges and future directions in targeting macrophages have also been discussed. Overall, our review highlights the significance of this versatile immune cell in human health and disease, which is expected to inform future research and clinical practice.
Collapse
Affiliation(s)
- Fan Guan
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Ruixuan Wang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zhenjie Yi
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Wanyao Liu
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Yao Xie
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Zaoqu Liu
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhiwei Xia
- Department of Neurology, Hunan Aerospace Hospital, Hunan Normal University, Changsha, China.
| | - Hao Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China.
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
22
|
Wei X, Qian W, Narasimhan H, Chan T, Liu X, Arish M, Young S, Li C, Cheon IS, Yu Q, Almeida-Santos G, Zhao XY, Yeatts EV, Spear OJ, Yi M, Parimon T, Fang Y, Hahn YS, Bullock TNJ, Somerville LA, Kaplan MH, Sperling AI, Shim YM, Vassallo R, Chen P, Ewald SE, Roden AC, Que J, Jiang D, Sun J. Macrophage peroxisomes guide alveolar regeneration and limit SARS-CoV-2 tissue sequelae. Science 2025; 387:eadq2509. [PMID: 40048515 DOI: 10.1126/science.adq2509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 10/20/2024] [Accepted: 12/19/2024] [Indexed: 04/23/2025]
Abstract
Peroxisomes are vital but often overlooked metabolic organelles. We found that excessive interferon signaling remodeled macrophage peroxisomes. This loss of peroxisomes impaired inflammation resolution and lung repair during severe respiratory viral infections. Peroxisomes were found to modulate lipid metabolism and mitochondrial health in a macrophage type-specific manner and enhanced alveolar macrophage-mediated tissue repair and alveolar regeneration after viral infection. Peroxisomes also prevented excessive macrophage inflammasome activation and IL-1β release, limiting accumulation of KRT8high dysplastic epithelial progenitors following viral injury. Pharmacologically enhancing peroxisome biogenesis mitigated both acute symptoms and post-acute sequelae of COVID-19 (PASC) in animal models. Thus, macrophage peroxisome dysfunction contributes to chronic lung pathology and fibrosis after severe acute respiratory syndrome coronavirus 2 infection.
Collapse
Affiliation(s)
- Xiaoqin Wei
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, USA
- Division of Infectious Disease and International Health, Department of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Wei Qian
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, USA
- Division of Infectious Disease and International Health, Department of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Harish Narasimhan
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, USA
- Division of Infectious Disease and International Health, Department of Medicine, University of Virginia, Charlottesville, VA, USA
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA, USA
| | - Ting Chan
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, USA
- Division of Infectious Disease and International Health, Department of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Xue Liu
- Women's Guild Lung Institute, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Mohd Arish
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, USA
- Division of Infectious Disease and International Health, Department of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Samuel Young
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, USA
- Division of Infectious Disease and International Health, Department of Medicine, University of Virginia, Charlottesville, VA, USA
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA, USA
| | - Chaofan Li
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, USA
- Division of Infectious Disease and International Health, Department of Medicine, University of Virginia, Charlottesville, VA, USA
| | - In Su Cheon
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, USA
- Division of Infectious Disease and International Health, Department of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Qing Yu
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, USA
- Division of Infectious Disease and International Health, Department of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Gislane Almeida-Santos
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, USA
- Division of Infectious Disease and International Health, Department of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Xiao-Yu Zhao
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, USA
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA, USA
| | - Eric V Yeatts
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, USA
- Division of Infectious Disease and International Health, Department of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Olivia J Spear
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, USA
- Division of Infectious Disease and International Health, Department of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Megan Yi
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, USA
- Division of Infectious Disease and International Health, Department of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Tanyalak Parimon
- Women's Guild Lung Institute, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Yinshan Fang
- Columbia Center for Human Development, Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | - Young S Hahn
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, USA
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA, USA
| | - Timothy N J Bullock
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, USA
- Department of Pathology, University of Virginia, Charlottesville, VA, USA
| | - Lindsay A Somerville
- Division of Pulmonary Medicine and Critical Care Medicine, Department of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Mark H Kaplan
- Department of Microbiology and Immunology, Indiana University of School of Medicine, Indianapolis, IN, USA
| | - Anne I Sperling
- Division of Pulmonary Medicine and Critical Care Medicine, Department of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Yun Michael Shim
- Division of Pulmonary Medicine and Critical Care Medicine, Department of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Robert Vassallo
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Peter Chen
- Women's Guild Lung Institute, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Sarah E Ewald
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, USA
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA, USA
| | - Anja C Roden
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Jianwen Que
- Columbia Center for Human Development, Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | - Dianhua Jiang
- Women's Guild Lung Institute, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Jie Sun
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, USA
- Division of Infectious Disease and International Health, Department of Medicine, University of Virginia, Charlottesville, VA, USA
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
23
|
Chen Q, Chen W, Zhang B, Xue L, Li F, Zhang L, Tong H, Zhu Q. Hesperetin mitigates adipose tissue inflammation to improve obesity-associated metabolic health. Int Immunopharmacol 2025; 149:114211. [PMID: 39929097 DOI: 10.1016/j.intimp.2025.114211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 01/26/2025] [Accepted: 01/31/2025] [Indexed: 02/22/2025]
Abstract
Metabolically unhealthy obesity (MUO) poses significant health risks, including increased susceptibility to type 2 diabetes and cardiovascular diseases. Hesperetin is a key bioactive compound found in citrus fruits. Previous studies have shown that hesperetin can correct metabolic abnormalities and mitigate the progression of various metabolic disorders, but the underlying mechanisms remain unclear. Here, we explored the impact of hesperetin on MUO using ob/ob mice and investigated its potential pharmacological mechanisms. The present data indicated that administration of hesperetin for 12 weeks led to notable improvements in metabolic parameters, including reduced fasting blood glucose, fasting insulin levels, and the HOMA-IR index in ob/ob mice. Glucose and insulin tolerance tests demonstrated that hesperetin effectively enhanced insulin sensitivity, with high-dose effects comparable to metformin. Hesperetin treatment decreased inguinal white adipose tissue (iWAT) weight and improved insulin signaling by increasing AKT phosphorylation. Additionally, it reduced the expression of pro-inflammatory cytokines (Il-6 and Il-1β), chemokine Ccl2 and its receptor Ccr2, and macrophage activation markers Nos2 and Ptgs2 within iWAT of ob/ob mice, likely by inhibiting NF-κB activation and macrophage-mediated inflammation. In vitro studies further confirmed hesperetin's anti-inflammatory effects in LPS-stimulated macrophages, where it suppressed cytokine production and NF-κB signaling. Hesperetin also impaired CCL2-induced macrophage chemotaxis, reducing migration velocity and distance. Mechanistically, hesperetin directly interacts with and inhibits IKKβ kinase activity by binding to key residues (LEU21, VAL465, CYS99, and GLU97) and stabilizing the complex, as demonstrated by molecular docking and molecular dynamics simulations. These findings underscore hesperetin's therapeutic potential in mitigating metabolically unhealthy obesity, obesity-induced insulin resistance, and inflammation through direct modulation of the IKKβ and NF-κB pathways.
Collapse
Affiliation(s)
- Qiu Chen
- Department of Endocrinology, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Wenjun Chen
- Department of Gastroenterology, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, China
| | - Beining Zhang
- First College of Clinical Medicine, Wenzhou Medical University, Wenzhou 325000, China
| | - Liwei Xue
- Department of Gastroenterology, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, China
| | - Fang Li
- College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China
| | - Lin Zhang
- College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China
| | - Haibin Tong
- College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China; State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Beijing 100700, China.
| | - Qihan Zhu
- Department of Endocrinology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China; Wenzhou Key Laboratory of Diabetes Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China.
| |
Collapse
|
24
|
Chen J, Xu D, Chen B, Jian S, Chen T, Wang W, Ma Y, Zhang J, Li K, Cai W, Xiao M. Senescent macrophages trigger a pro-inflammatory program and promote the progression of rheumatoid arthritis. Int Immunopharmacol 2025; 149:114164. [PMID: 39904033 DOI: 10.1016/j.intimp.2025.114164] [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: 09/11/2024] [Revised: 12/27/2024] [Accepted: 01/22/2025] [Indexed: 02/06/2025]
Abstract
This study explores the complex mechanisms between Rheumatoid Arthritis (RA) and cellular senescence, with a focus on the role of four key genes (MMP1, CCL7, CXCL1, HK3) identified through transcriptome analysis in the GEO database. These genes are closely related to IL-17 signalling and the pathogenesis of RA. In a macrophage senescence model induced by hydrogen peroxide (H2O2) and bleomycin (BLM), quantitative real-time PCR (qRT-PCR) and Western blot confirmed the significant upregulation of these genes and an increase in the secretion of the cytokine IL-17, which promotes an inflammatory environment for the polarization of macrophages to M1. When co-cultured with mouse synovial fibroblasts (MSF), MSF showed enhanced vitality and increased invasiveness, indicating a key role for these genes in the progression of RA. Additionally, HK3, less reported in RA, when its expression is knocking down, lactate secretion and lactylation modification at lysine 14 of histone H3 in macrophages were reduced, leading to a change in macrophage polarity. The study concludes that the altered polarity of senescent macrophages drives the proliferation and invasion of MSF, significantly promoting the development of RA and providing insights into the pathophysiology of RA.
Collapse
Affiliation(s)
- Jiayao Chen
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, School of Basic Medicine and Life Sciences, Hainan Medical University, Haikou 571199 Hainan, China
| | - Dahua Xu
- College of Biomedical Information and Engineering, Hainan Medical University, Haikou 571199 Hainan, China; Hainan Engineering Research Center for Health Big Data, Hainan Medical University, Haikou 571199 Hainan, China
| | - Bocen Chen
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, School of Basic Medicine and Life Sciences, Hainan Medical University, Haikou 571199 Hainan, China
| | - Shaoqin Jian
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, School of Basic Medicine and Life Sciences, Hainan Medical University, Haikou 571199 Hainan, China
| | - Tong Chen
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, School of Basic Medicine and Life Sciences, Hainan Medical University, Haikou 571199 Hainan, China
| | - Wenguang Wang
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, Department of Reproductive Medicine, Hainan Provincial Clinical Research Center for Thalassemia, Key Laboratory of Reproductive Health Diseases Research and Translation (Hainan Medical University), Ministry of Education, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou 571199 Hainan, China
| | - Yingjie Ma
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, Department of Reproductive Medicine, Hainan Provincial Clinical Research Center for Thalassemia, Key Laboratory of Reproductive Health Diseases Research and Translation (Hainan Medical University), Ministry of Education, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou 571199 Hainan, China
| | - Jing Zhang
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, School of Basic Medicine and Life Sciences, Hainan Medical University, Haikou 571199 Hainan, China
| | - Kongning Li
- College of Biomedical Information and Engineering, Hainan Medical University, Haikou 571199 Hainan, China; Hainan Engineering Research Center for Health Big Data, Hainan Medical University, Haikou 571199 Hainan, China.
| | - Wangwei Cai
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, School of Basic Medicine and Life Sciences, Hainan Medical University, Haikou 571199 Hainan, China.
| | - Man Xiao
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, School of Basic Medicine and Life Sciences, Hainan Medical University, Haikou 571199 Hainan, China.
| |
Collapse
|
25
|
Njoku GC, Forkan CP, Soltysik FM, Nejsum PL, Pociot F, Yarani R. Unleashing the potential of extracellular vesicles for ulcerative colitis and Crohn's disease therapy. Bioact Mater 2025; 45:41-57. [PMID: 39610953 PMCID: PMC11602541 DOI: 10.1016/j.bioactmat.2024.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 10/28/2024] [Accepted: 11/05/2024] [Indexed: 11/30/2024] Open
Abstract
Image 1.
Collapse
Affiliation(s)
- George Chigozie Njoku
- Translational Type 1 Diabetes Research, Department of Clinical and Translational Research, Steno Diabetes Center Copenhagen, Herlev, Denmark
- Department of Medical Biotechnology, University of Naples Federico II, Naples, Italy
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, USA
| | - Cathal Patrick Forkan
- Translational Type 1 Diabetes Research, Department of Clinical and Translational Research, Steno Diabetes Center Copenhagen, Herlev, Denmark
- Department of Pharmacy, Université Grenoble Alpes, France
| | - Fumie Mitani Soltysik
- Translational Type 1 Diabetes Research, Department of Clinical and Translational Research, Steno Diabetes Center Copenhagen, Herlev, Denmark
| | - Peter Lindberg Nejsum
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| | - Flemming Pociot
- Translational Type 1 Diabetes Research, Department of Clinical and Translational Research, Steno Diabetes Center Copenhagen, Herlev, Denmark
| | - Reza Yarani
- Translational Type 1 Diabetes Research, Department of Clinical and Translational Research, Steno Diabetes Center Copenhagen, Herlev, Denmark
| |
Collapse
|
26
|
Zhao B, Li M, Zhang H, Wang J, Zhao W, Yang Y, Usman M, Loor JJ, Xu C. M1 polarization of hepatic macrophages in cows with subclinical ketosis is an important cause of liver injury. J Dairy Sci 2025; 108:2933-2946. [PMID: 39647630 DOI: 10.3168/jds.2024-25500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 11/12/2024] [Indexed: 12/10/2024]
Abstract
Subclinical ketosis (SCK) is highly prevalent and easily overlooked, with insidious and slow progression of hepatic injury, often characterized by an imbalance in immune homeostasis. In nonruminants, macrophage polarization plays an important regulatory role in hepatic lipid accumulation, fibrosis, and inflammatory processes. Thus, we aimed to investigate the status of hepatic macrophage polarization in SCK cows and to corroborate its association with liver injury and inflammation. Twelve Holstein dairy cows (parity 2-4) were selected, and liver biopsy and blood were collected on the second week postpartum (10-14 d DIM). On the basis of serum beta-hydroxybutyric acid (BHBA) concentrations, selected cows were categorized into healthy (n = 6; BHBA <1.0 mM) and SCK (n = 6; 1.2 mM ≤ BHBA < 3.0 mM) groups. Serum biochemical parameters were measured using an automatic biochemical analyzer, which indicated higher serum levels of BHBA and nonesterified fatty acids and an upregulation of liver injury indicators (aspartate aminotransferase [AST], alanine aminotransferase [ALT], total protein, globulin) in SCK cows compared with healthy cows. The ELISA assays revealed that SCK cows displayed systemic low-grade inflammation, as demonstrated by increased serum levels of haptoglobin, serum amyloid A, TGF-β, IFN-γ, and IL-1β. Liver biopsies revealed pathological histological alterations, hepatic inflammation, and macrophage polarization status. Oil Red staining indicated steatosis, whereas Sirius red staining demonstrated mild extracellular matrix deposition in the liver of SCK cows. The expression of inflammatory response-related proteins (TLR4, p-NFκB, p-I-κB, NLRP3, and Caspase 1) was elevated in the liver of SCK cows, with the increased mean fluorescence intensity of NFκB further confirming the activation of the inflammatory pathway. Furthermore, the levels of pro-inflammatory factors, TNF-α and IFN-γ, were elevated in the tissue homogenate. Macrophage phenotypic changes in SCK cows were further explored based on the results of liver injury and inflammation. Compared with healthy cows, the protein and mRNA abundance of the macrophage marker CD68 in the liver of SCK cows was higher, along with an increased mean fluorescence intensity of CD68. The SCK cows also exhibited reduced mRNA expression of the Kupffer cell marker CLEC4F and elevated chemokine levels (CXCL1 and CCL2). As evidenced by greater protein and mRNA abundance of macrophage M1 polarization markers (iNOS, IL-1β, CD86, IL-6, IL-12b, and CCL3), higher fluorescence intensity of iNOS and CD86, and an increased number of CD68+/CD86+-positive cells observed via immunofluorescence, the macrophage polarization phenotype in the liver of SCK cows was predominantly M1. In contrast, the protein and mRNA abundances of M2 polarization markers (CD206, IL-10, and Arg1) were lower in SCK cows, accompanied by a reduced fluorescence intensity of CD206 and a lower number of CD68+/CD206+-positive cells. Overall, the present study revealed that the number of macrophages in liver is enhanced during subclinical ketosis and is dominated by pro-inflammatory macrophages (M1 macrophages). This could partly explain the increased risk of steatosis, fibrosis, and inflammatory response processes in these cows.
Collapse
Affiliation(s)
- Bichen Zhao
- College of Veterinary Medicine, China Agricultural University, 100193, Beijing, China
| | - Ming Li
- College of Veterinary Medicine, China Agricultural University, 100193, Beijing, China
| | - Huijing Zhang
- College of Veterinary Medicine, China Agricultural University, 100193, Beijing, China
| | - Jingyi Wang
- College of Veterinary Medicine, China Agricultural University, 100193, Beijing, China
| | - Wanli Zhao
- College of Veterinary Medicine, China Agricultural University, 100193, Beijing, China
| | - Yue Yang
- College of Veterinary Medicine, China Agricultural University, 100193, Beijing, China
| | - Muhammad Usman
- Mammalian NutriPhysio Genomics, Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana, IL 61801
| | - Juan J Loor
- Mammalian NutriPhysio Genomics, Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana, IL 61801
| | - Chuang Xu
- College of Veterinary Medicine, China Agricultural University, 100193, Beijing, China.
| |
Collapse
|
27
|
Zhao W, Jia Z, Han J, Sun X. Boswellia Extract Promotes Healing and Resolving Inflammation in Oral Ulcers of Rat. J Oral Pathol Med 2025; 54:131-140. [PMID: 39871413 DOI: 10.1111/jop.13609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 11/11/2024] [Accepted: 12/10/2024] [Indexed: 01/29/2025]
Abstract
BACKGROUND Recurrent aphthous ulcers significantly impact patients' quality of life due to their painful and recurrent nature, necessitating more effective treatments. This study explores the therapeutic potential of Boswellia to treat recurrent aphthous ulcers by its anti-inflammatory and healing promotion effect in a rat oral ulcer model. METHODS Network pharmacology techniques were employed to elucidate Boswellia's active components and potential targets. Intersecting targets of Boswellia and oral ulcer-related genes were screened for protein-protein interaction network analysis and functional enrichment. An oral ulcer model in rats was used and rats were treated with Boswellia extract. The healing process was monitored by measuring the ulcer area and body weight changes. Histological analysis was performed, and the role of Boswellia in macrophage polarization was investigated through gene expression analysis and protein array tests. The underlying mechanism involving PPARγ activation was also explored. RESULTS Network pharmacology analysis revealed Boswellia's interaction with key genes and pathways associated with inflammation and lipid metabolism, such as MAPK3, PPARG, and PTGS2. Boswellia extract significantly accelerated oral ulcer healing and recovered weight loss in rats. Histological examinations revealed reduced tissue swelling and inflammatory cell infiltration in treated groups. Furthermore, Boswellia extract decreased infiltration of M1 macrophage presence while increasing M2 macrophage, indicating an inflammation-resolving effect. In vitro studies showed that Boswellia extract enhanced M2-related gene expression and decreased pro-inflammatory cytokines, which is PPARγ dependent. CONCLUSION Boswellia extract promotes oral ulcer healing and resolves inflammation, primarily through the modulation of macrophage polarization via PPARγ activation.
Collapse
Affiliation(s)
- Wei Zhao
- Department of Oral Medicine, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Zhuoqun Jia
- Department of Oral Medicine, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Jiao Han
- Department of Oral Medicine, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Xiaojun Sun
- Department of Oral Medicine, First Hospital of Shanxi Medical University, Taiyuan, China
| |
Collapse
|
28
|
Rajput S, Malviya R, Srivastava S, Ahmad I, Rab SO, Uniyal P. Cardiovascular disease and thrombosis: Intersections with the immune system, inflammation, and the coagulation system. ANNALES PHARMACEUTIQUES FRANÇAISES 2025; 83:228-250. [PMID: 39159826 DOI: 10.1016/j.pharma.2024.08.005] [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: 01/11/2024] [Revised: 08/06/2024] [Accepted: 08/13/2024] [Indexed: 08/21/2024]
Abstract
The coagulation and immune system, both essential physiological systems in the human body, are intricately interconnected and play a critical role in determining the overall health of patients. These systems collaborate via various shared regulatory pathways, such as the Tissue Factor (TF) Pathway. Immunological cells that express TF and generate pro-inflammatory cytokines have the ability to affect coagulation. Conversely, coagulation factors and processes have a reciprocal effect on immunological responses by stimulating immune cells and regulating their functions. These interconnected pathways play a role in both preserving well-being and contributing to a range of pathological disorders. The close relationship between blood clotting and inflammation in the development of vascular disease has become a central focus of clinical study. This research specifically examines the crucial elements of this interaction within the contexts of cardiovascular disease and acute coronary syndrome. Tissue factor, the primary trigger of the extrinsic coagulation pathway, has a crucial function by inducing a proinflammatory reaction through the activation of coagulation factors. This, in turn, initiates coagulation and subsequent cellular signalling pathways. Protease-activated receptors establish the molecular connection between coagulation and inflammation by interacting with activated clotting factors II, X, and VII. Thrombosis, a condition characterised by the formation of blood clots, is the most dreaded consequence of cardiovascular disorders and a leading cause of death globally. Consequently, it poses a significant challenge to healthcare systems. Antithrombotic treatments efficiently target platelets and the coagulation cascade, but they come with the inherent danger of causing bleeding. Furthermore, antithrombotics are unable to fully eliminate thrombotic events, highlighting a treatment deficiency caused by a third mechanism that has not yet been sufficiently addressed, namely inflammation. Understanding these connections may aid in the development of novel approaches to mitigate the harmful mutual exacerbation of inflammation and coagulation. Gaining a comprehensive understanding of the intricate interaction among these systems is crucial for the management of diseases and the creation of efficacious remedies. Through the examination of these prevalent regulatory systems, we can discover novel therapeutic approaches that specifically target these complex illnesses. This paper provides a thorough examination of the reciprocal relationship between the coagulation and immune systems, emphasising its importance in maintaining health and understanding disease processes. This review examines the interplay between inflammation and thrombosis and its role in the development of thrombotic disorders.
Collapse
Affiliation(s)
- Shivam Rajput
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, U.P., India
| | - Rishabha Malviya
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, U.P., India.
| | - Saurabh Srivastava
- School of Pharmacy, KPJ Healthcare University College (KPJUC), Nilai, Malaysia
| | - Irfan Ahmad
- Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia
| | - Safia Obaidur Rab
- Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia
| | - Prerna Uniyal
- School of Pharmacy, Graphic Era Hill University, Dehradun, India
| |
Collapse
|
29
|
Niu S, Li M, Wang J, Zhong P, Wen X, Huang F, Yin L, Liao Y, Zhou J. Identify the potential target of efferocytosis in knee osteoarthritis synovial tissue: a bioinformatics and machine learning-based study. Front Immunol 2025; 16:1550794. [PMID: 40083558 PMCID: PMC11903261 DOI: 10.3389/fimmu.2025.1550794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Accepted: 02/11/2025] [Indexed: 03/16/2025] Open
Abstract
Introduction Knee osteoarthritis (KOA) is a degenerative joint disease characterized by the progressive deterioration of cartilage and synovial inflammation. A critical mechanism in the pathogenesis of KOA is impaired efferocytosis in synovial tissue. The present study aimed to identify and validate key efferocytosis-related genes (EFRGs) in KOA synovial tissue by using comprehensive bioinformatics and machine learning approaches. Methods We integrated three datasets (GSE55235, GSE55457, and GSE12021) from the Gene Expression Omnibus database to screen differentially expressed genes (DEGs) associated with efferocytosis and performed weighted gene co-expression network analysis. Subsequently, we utilized univariate logistic regression analysis, least absolute shrinkage and selection operator regression, support vector machine, and random forest algorithms to further refine these genes. The results were then inputted into multivariate logistic regression analysis to construct a diagnostic nomogram. Public datasets and quantitative real-time PCR experiments were employed for validation. Additionally, immune infiltration analysis was conducted with CIBERSORT using the combined datasets. Results Analysis of the intersection between DEGs and EFRGs identified 12 KOA-related efferocytosis DEGs. Further refinement through machine learning algorithms and multivariate logistic regression revealed UCP2, CX3CR1, and CEBPB as hub genes. Immune infiltration analysis demonstrated significant correlations between immune cell components and the expression levels of these hub genes. Validation using independent datasets and experimental approaches confirmed the robustness of these findings. Conclusions This study successfully identified three hub genes (UCP2, CX3CR1, and CEBPB) with significant expression alterations in KOA, demonstrating high diagnostic potential and close associations with impaired efferocytosis. These targets may modulate synovial efferocytosis-related immune processes, offering novel therapeutic avenues for KOA intervention.
Collapse
Affiliation(s)
- Shangbo Niu
- Rehabilitation Medicine Center Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, Hunan, China
- Department of Rehabilitation, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Rehabilitation Laboratory, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Mengmeng Li
- Rehabilitation Medicine Center Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, Hunan, China
- Department of Rehabilitation, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Rehabilitation Laboratory, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Jinling Wang
- Rehabilitation Medicine Center Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, Hunan, China
- Department of Rehabilitation, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Rehabilitation Laboratory, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Peirui Zhong
- Rehabilitation Medicine Center Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, Hunan, China
- Department of Rehabilitation, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Rehabilitation Laboratory, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Xing Wen
- Rehabilitation Medicine Center Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, Hunan, China
- Department of Rehabilitation, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Rehabilitation Laboratory, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Fujin Huang
- Rehabilitation Medicine Center Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, Hunan, China
- Department of Rehabilitation, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Rehabilitation Laboratory, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Linwei Yin
- Rehabilitation Medicine Center Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, Hunan, China
- Department of Rehabilitation, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Rehabilitation Laboratory, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Yang Liao
- Rehabilitation Medicine Center Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, Hunan, China
- Department of Rehabilitation, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Rehabilitation Laboratory, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Jun Zhou
- Rehabilitation Medicine Center Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, Hunan, China
- Department of Rehabilitation, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Rehabilitation Laboratory, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| |
Collapse
|
30
|
Fei C, Chen Y, Tan R, Yang X, Wu G, Li C, Shi J, Le S, Yang W, Xu J, Wang L, Zhang Z. Single-cell multi-omics analysis identifies SPP1 + macrophages as key drivers of ferroptosis-mediated fibrosis in ligamentum flavum hypertrophy. Biomark Res 2025; 13:33. [PMID: 40001138 PMCID: PMC11863437 DOI: 10.1186/s40364-025-00746-6] [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: 11/08/2024] [Accepted: 02/11/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND Ligamentum flavum hypertrophy (LFH) is a primary contributor to lumbar spinal stenosis. However, a thorough understanding of the cellular and molecular mechanisms driving LFH fibrotic progression remains incomplete. METHODS Single-cell RNA sequencing (scRNA-seq) was performed to construct the single-cell map of human ligamentum flavum (LF) samples. An integrated multi-omics approach, encompassing scRNA-seq, bulk RNA sequencing (bulk RNA-seq), and Mendelian randomization (MR), was applied to conduct comprehensive functional analysis. Clinical tissue specimens and animal models were employed to further confirm the multi-omics findings. RESULTS ScRNA-seq provided a single-cell level view of the fibrotic microenvironment in LF, revealing significantly increased proportions of fibroblasts, myofibroblasts, and macrophages in LFH. Using transmission electron microscopy, single-cell gene set scoring, and MR analysis, ferroptosis was identified as a critical risk factor and pathway within LFH. Subcluster analysis of fibroblasts revealed functional heterogeneity among distinct subpopulations, highlighting the functional characteristics and the metabolic dynamics of fibroblast with a high ferroptosis score (High Ferro-score FB). The quantification of gene expression at single-cell level revealed that ferroptosis increased along with fibrosis in LFH specimens, a finding further validated in both human and mice tissue sections. Consistently, bulk RNA-seq confirmed increased proportions of fibroblasts and macrophages in LFH specimens, underscoring a strong correlation between these cell types through Spearman correlation analysis. Notably, subcluster analysis of the mononuclear phagocytes identified a specific subset of SPP1+ macrophages (SPP1+ Mac) enriched in LFH, which exhibited activation of fibrosis and ferroptosis-related metabolic pathways. Cell-cell communication analysis highlighted that SPP1+ Mac exhibited the strongest outgoing and incoming interactions among mononuclear phagocytes in the LFH microenvironment. Ligand-receptor analysis further revealed that the SPP1-CD44 axis could serve as a key mediator regulating the activity of High Ferro-score FB. Multiplex immunofluorescence confirmed substantial Collagen I deposition and reduced Ferritin Light Chain expression in regions with SPP1-CD44 co-localization in LFH specimens. CONCLUSIONS Our findings indicated that SPP1+ Mac may contribute to LFH fibrosis by regulating ferroptosis in High Ferro-score FB through the SPP1-CD44 axis. This study enhances our understanding of the cellular and molecular mechanisms underlying LFH progression, potentially improving early diagnostic strategies and identifying new therapeutic targets.
Collapse
Affiliation(s)
- Chengshuo Fei
- Division of Spine Surgery, Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yanlin Chen
- Division of Spine Surgery, Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Ruiqian Tan
- Division of Spine Surgery, Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Xinxing Yang
- Division of Spine Surgery, Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Guanda Wu
- Division of Spine Surgery, Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Chenglong Li
- Division of Spine Surgery, Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Jiawei Shi
- Division of Spine Surgery, Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Shiyong Le
- Division of Spine Surgery, Department of Orthopaedics, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510630, China
| | - Wenjie Yang
- Division of Spine Surgery, Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Jiajia Xu
- Division of Spine Surgery, Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Liang Wang
- Division of Spine Surgery, Department of Orthopaedics, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510630, China.
| | - Zhongmin Zhang
- Division of Spine Surgery, Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
31
|
Wu R, Yang H, Liu C. IFIT3: a crucial mediator in innate immunity and tumor progression with therapeutic implications. Front Immunol 2025; 16:1515718. [PMID: 40061935 PMCID: PMC11885914 DOI: 10.3389/fimmu.2025.1515718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 01/30/2025] [Indexed: 05/13/2025] Open
Abstract
Interferon-Induced Protein with Tetratricopeptide Repeats 3 (IFIT3) plays a dual role in innate immunity and tumor immunity, functioning as both a viral defense molecule and a regulator of tumor progression. This review explores the mechanisms through which IFIT3 modulates immune responses, including interferon signaling, RIG-I-like receptors, and the NF-κB pathway. IFIT3 facilitates immune evasion and promotes inflammation-mediated tumor growth by regulating immune checkpoints and the tumor microenvironment, its emerging role as a target for cancer immunotherapy opens new avenues for therapeutic strategies. Finally, this paper underscores IFIT3's potential clinical applications in the modulation of tumor immunity, highlighting the need for further research on IFIT3-targeted therapies.
Collapse
Affiliation(s)
- Rihan Wu
- Department of Radiation Oncology, Peking University Cancer Hospital (Inner Mongolia Campus) & Affiliated Cancer Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region, China
- Graduate School, Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region, China
| | - Hao Yang
- Department of Radiation Oncology, Peking University Cancer Hospital (Inner Mongolia Campus) & Affiliated Cancer Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region, China
- Graduate School, Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region, China
| | - Chunlei Liu
- Translational Medicine Research Center, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
32
|
Jin C, Wu P, Wu W, Chen W, Liu W, Zhu Y, Wu Q, Chen B, Ji C, Qian H. Therapeutic role of hucMSC-sEV-enriched miR-13896 in cisplatin-induced acute kidney injury through M2 macrophage polarization. Cell Biol Toxicol 2025; 41:50. [PMID: 39992453 PMCID: PMC11850457 DOI: 10.1007/s10565-025-09998-2] [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: 10/02/2024] [Accepted: 02/11/2025] [Indexed: 02/25/2025]
Abstract
Human umbilical cord mesenchymal stem cell-derived small extracellular vesicles (hucMSC-sEV) have recently garnered attention as a potential therapeutic approach for kidney diseases with anti-inflammatory effects. Infiltrated macrophages play an important role in facilitating tissue regeneration. However, the intricate regulatory effects of hucMSC-sEV on macrophages during cisplatin-induced acute kidney injury (AKI) remain unknown. In this study, we uncovered that hucMSC-sEV exhibited potent anti-inflammation and effectively inhibited the polarization of M1 phenotype macrophages. Mechanically, miRNA sequencing analysis and qRT-PCR indicated that a novel miRNA, named miR-13896, was enriched in hucMSC-sEV. When transfected with miR-13896 mimic, macrophages displayed M2 phenotype with elevated levels of Arg1 and IL-10, while miR-13896 inhibitor promoted M1 phenotype. Furthermore, we firstly established that miR-13896 repressed Tradd expression by targeting its 3' untranslated region and subsequently inhibited NF-κB signaling pathway in macrophages. Additionally, to improve therapeutic effects, hucMSC-sEV were engineered with elevated levels of miR-13896 through electroporation, which resulted in promoting M2 phenotype macrophages, inhibiting inflammatory factors, and enhancing kidney repair. Conclusively, our findings provide novel insights into the mechanisms underlying the effects of hucMSC-sEV on macrophages and AKI, while also highlighting electroporation as a promising strategy for treating cisplatin-induced AKI.
Collapse
Affiliation(s)
- Can Jin
- Institute of Translational Medicine of Jiangsu University, Department of Urology, Affiliated Hospital of Jiangsu University, 438 Jiefang Road, Zhenjiang, 212001, Jiangsu, China
- Key Laboratory of Laboratory Medicine of Jiangsu Province, Department of Laboratory Medicine, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, China
| | - Peipei Wu
- Key Laboratory of Laboratory Medicine of Jiangsu Province, Department of Laboratory Medicine, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, China
- Department of Clinical Laboratory, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Wei Wu
- Department of Emergency Surgery, Qinghai Provincial People's Hospital, 2 Gonghe Road, Xining, 810007, Qinghai, China
| | - Wenya Chen
- Key Laboratory of Laboratory Medicine of Jiangsu Province, Department of Laboratory Medicine, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, China
| | - Wanzhu Liu
- Department of Emergency Medicine, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, 212002, Jiangsu, China
| | - Yuan Zhu
- Department of Emergency Medicine, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, 212002, Jiangsu, China
| | - QiShun Wu
- Institute of Translational Medicine of Jiangsu University, Department of Urology, Affiliated Hospital of Jiangsu University, 438 Jiefang Road, Zhenjiang, 212001, Jiangsu, China
| | - Binghai Chen
- Institute of Translational Medicine of Jiangsu University, Department of Urology, Affiliated Hospital of Jiangsu University, 438 Jiefang Road, Zhenjiang, 212001, Jiangsu, China.
| | - Cheng Ji
- Key Laboratory of Laboratory Medicine of Jiangsu Province, Department of Laboratory Medicine, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, China.
| | - Hui Qian
- Institute of Translational Medicine of Jiangsu University, Department of Urology, Affiliated Hospital of Jiangsu University, 438 Jiefang Road, Zhenjiang, 212001, Jiangsu, China.
- Key Laboratory of Laboratory Medicine of Jiangsu Province, Department of Laboratory Medicine, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, China.
- NHC Key Laboratory of Medical Embryogenesis and Developmental Molecular Biology & Shanghai Key Laboratory of Embryo and Reproduction Engineering, Shanghai, 200040, China.
- Institute of Translational Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, China.
| |
Collapse
|
33
|
Cheng Y, Dong X, Shi J, Wu G, Tao P, Ren N, Zhao Y, Li F, Wang Z. Immunomodulation with M2 macrophage-derived extracellular vesicles for enhanced titanium implant osseointegration under diabetic conditions. Mater Today Bio 2025; 30:101385. [PMID: 39742145 PMCID: PMC11683253 DOI: 10.1016/j.mtbio.2024.101385] [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: 08/03/2024] [Revised: 11/28/2024] [Accepted: 12/04/2024] [Indexed: 01/03/2025] Open
Abstract
M2 macrophage-derived extracellular vesicles (M2-EVs) demonstrate the capacity to reduce pro-inflammatory M1 macrophage formation, thereby restoring the M1-M2 macrophage balance and promoting immunoregulation. However, the efficacy of M2-EVs in regulating macrophage polarization and subsequently enhancing osseointegration around titanium (Ti) implants in patients with diabetes mellitus (DM) remains to be elucidated. In this study, Ti implants were coated with polydopamine to facilitate M2-EVs adherence. In vitro experiment results demonstrated that M2-EVs could carry miR-23a-3p, inhibiting NOD-like receptor protein3(NLRP3) inflammasome activation in M1 macrophage and reducing the levels of inflammatory cytokines such as IL-1β by targeting NEK7. This improved the M1-M2 macrophage balance and enhanced mineralization on the Ti implant surfaces. The in vivo experiment results demonstrated that in diabetic conditions, the nanocoated M2-EVs significantly promoted high-quality bone deposition around the Ti implants. The current results provide a novel perspective for simple and effective decoration of M2-EVs on Ti implants; clinically, the method may afford osteoimmunomodulatory effects enhancing implant osseointegration in patients with DM.
Collapse
Affiliation(s)
- Yuzhao Cheng
- The Stomatology Department of Shanxi Provincial People Hospital, Shanxi Medical University, Taiyuan, Shanxi, 030001, China
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, China
| | - Xin Dong
- Department of Orthopedic Surgery, Tangdu Hospital, Air Force Military Medical University, Xi'an, Shaanxi Province, China
| | - Jing Shi
- The Stomatology Department of Shanxi Provincial People Hospital, Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Guangsheng Wu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, China
- Department of Stomatology, Qingdao Special Servicemen Recuperation Center of PLA Navy, No.18 Yueyang Road, Qingdao, 266071, China
| | - Pei Tao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, China
- College of Chemistry and Bio-engineering, Yichun University, Yichun, Jiangxi, 336000, China
| | - Nan Ren
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, China
| | - Yimin Zhao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, China
| | - Fenglan Li
- The Stomatology Department of Shanxi Provincial People Hospital, Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Zhongshan Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, China
| |
Collapse
|
34
|
Li R, Li X, Zhang X, Yu J, Li Y, Ran S, Wang S, Luo Z, Zhao J, Hao Y, Zong J, Zheng K, Lai L, Zhang H, Huang P, Zhou C, Wu J, Ye W, Xia J. Macrophages in Cardiovascular Fibrosis: Novel Subpopulations, Molecular Mechanisms, and Therapeutic Targets. Can J Cardiol 2025; 41:309-322. [PMID: 39580052 DOI: 10.1016/j.cjca.2024.11.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 11/12/2024] [Accepted: 11/18/2024] [Indexed: 11/25/2024] Open
Abstract
Cardiovascular fibrosis is a common pathological process that contributes to the development and progression of various cardiovascular diseases. Despite being widely believed to be an irreversible and relentless process, preclinical models and clinical trials have shown that cardiovascular fibrosis is an extremely dynamic process. Additionally, as part of the innate immune system, macrophages are heterogeneous cells that are pivotal in tissue regeneration and fibrosis. They participate in fibroblast activation, extracellular matrix remodelling, and the regression of fibrosis. Although we have made some advances in understanding macrophages in cardiovascular fibrosis, a gap still remains between their identification and conversion into effective treatments. Moreover, the traditional M1-M2 paradigm faces many challenges because it does not sufficiently clarify macrophage diversity and their functions. Exploring novel macrophage-based therapies is urgent for cardiovascular fibrosis treatment. Single-cell techniques have shed light on identifying novel subpopulations that differ in function and molecular signature under steady-state and pathological conditions. In this review, we outline the developmental origins of macrophages, which underlie their functions; and recent technology development in the single-cell era. In addition, we describe the markers and mediators of the newly defined macrophage subpopulations and the molecular mechanisms involved to elucidate potential approaches for targeting macrophages in cardiovascular fibrosis.
Collapse
Affiliation(s)
- Ran Li
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaohan Li
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xi Zhang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jizhang Yu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuan Li
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shuan Ran
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Song Wang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zilong Luo
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiulu Zhao
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yanglin Hao
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Junjie Zong
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kexiao Zheng
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Longyong Lai
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Han Zhang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Pinyan Huang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Cheng Zhou
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jie Wu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Weicong Ye
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Jiahong Xia
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China.
| |
Collapse
|
35
|
Oh JH, Jeong MG, Lee S, Lim J, Kang J, Bae MA, Ahn JH, Hong JH, Hwang ES. SMEPPI: An indenone derivative that selectively inhibits M1 macrophage activation and enhances phagocytic activity. Biomed Pharmacother 2025; 183:117856. [PMID: 39813787 DOI: 10.1016/j.biopha.2025.117856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 01/03/2025] [Accepted: 01/13/2025] [Indexed: 01/18/2025] Open
Abstract
SMEPPI is a small molecule synthesized as a derivative of KR-62980 that has anti-diabetic and anti-inflammatory activities. Despite the established physiological effects of KR-62980, the effects and benefits of SMEPPI remain largely unexplored. This study investigated the immunomodulatory functions of SMEPPI on macrophages and inflammatory diseases. SMEPPI did not affect the differentiation and maturation of bone marrow-derived monocytes into macrophages, nor did it affect the proliferation of M1 or M2 macrophages. Although SMEPPI did not affect M2 macrophage polarization, it significantly inhibited IL-1β and IL-6 cytokine production in both M1 macrophages and activated RAW264.7 macrophages. Importantly, SMEPPI inhibited the expression and phosphorylation of NF-κB p65 through inhibition of Akt expression, preventing its translocation to the nucleus. It also promoted p65 degradation through the stimulation of the proteasomal degradation pathway by inducing the expression of proteasome-related genes, thereby inhibiting p65 transcriptional activity. SMEPPI also enhanced the expression of various molecules associated with macrophage phagocytosis, including CD68, CD33, and lectins, thereby increasing phagocytic activity. Moreover, SMEPPI mitigated lipopolysaccharides-induced acute lung injury by suppressing IL-1β and IL-6 production in M1 macrophages and reduced mortality related to severe lung injury. These findings indicate that SMEPPI effectively regulates inflammatory diseases by impeding p65-induced cytokine production and enhancement of phagocytosis by M1 macrophages.
Collapse
Affiliation(s)
- Ji Hyun Oh
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, South Korea
| | - Mi Gyeong Jeong
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, South Korea
| | - Soheun Lee
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, South Korea
| | - Jihae Lim
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, South Korea
| | - Jio Kang
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, South Korea
| | - Myung Ae Bae
- Drug Discovery Platform Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon 34114, South Korea
| | - Jin-Hee Ahn
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju 61005, South Korea
| | - Jeong-Ho Hong
- Division of Life Sciences, Korea University, Seoul 02841, South Korea
| | - Eun Sook Hwang
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, South Korea; Gradutate Program in Innovative Biomaterials Convergence, Ewha Womans University, Seoul 03760, South Korea.
| |
Collapse
|
36
|
Lv T, Fan R, Wu J, Gong H, Gao X, Liu X, Gong Y, Luo B, Zhang Y, Peng X, Liang G. Tumor-Associated Macrophages: Key Players in the Non-Small Cell Lung Cancer Tumor Microenvironment. Cancer Med 2025; 14:e70670. [PMID: 39927632 PMCID: PMC11808749 DOI: 10.1002/cam4.70670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 01/27/2025] [Accepted: 01/29/2025] [Indexed: 02/11/2025] Open
Abstract
BACKGROUND Lung cancer is among the most common and deadliest malignant tumors worldwide. It is often detected at late stages, resulting in unfavorable outcomes, with tumor cell heterogeneity and medication resistance. Tumor-associated macrophages are among the key cells contributing to cancer progression. They are categorized into two primary phenotypes: Proinflammatory (M1) and anti-inflammatory (M2) which are involved in the onset and progression of NSCLC. The role of common cytokines secreted by macrophages in the progression of lung cancer are described, and the effects of various substances such as RNA or protein on the differentiation and polarization of two phenotypes of macrophages are highlighted to characterize the impact of the immune state of tumors on therapeutic effect of treatments and patient prognosis. Researchers have primarily aimed to investigate innovative carriers and strategies based on macrophages to modify the tumor microenvironment. OBJECTIVES These approaches are often integrated with other treatments, particularly immunotherapy, to enhance therapeutic efficacy. METHODS A comprehensive review was carried out by systematically synthesizing existing literature on PubMed, using the combination of the keywords "TAMs", "NSCLC", "Drug resistance", and "therapy". The available studies were screened for selection based on quality and relevance. CONCLUSIONS TAMs promote tumor invasion, growth, and metastasis by promoting angiogenesis and EMT. In addition, they contribute to the development of drug resistance and the immunosuppressive microenvironment establishment. The immunosuppressive factors secreted by TAM can weaken the activity of immune cells, inhibit their killing effect on tumors, leading to immune suppression and hindering the effectiveness of treatment. Therefore, TAM is a key target for the development of cancer immunotherapy. Various strategies are being explored, including reducing the recruitment of TAMs and influencing their polarization to treat NSCLC. In addition, TAMs based treatment systems can achieve precise delivery of drugs or gene interfering molecules without causing side effects.
Collapse
Affiliation(s)
- Tongtong Lv
- Department of HematologyThe First Affiliated Hospital of Yangtze UniversityJingzhouChina
- Department of PathophysiologySchool of Basic Medicine, Health Science Center, Yangtze UniversityJingzhouChina
| | - Rui Fan
- Department of HematologyThe First Affiliated Hospital of Yangtze UniversityJingzhouChina
| | - Jiaqi Wu
- Department of HematologyThe First Affiliated Hospital of Yangtze UniversityJingzhouChina
- Department of PathophysiologySchool of Basic Medicine, Health Science Center, Yangtze UniversityJingzhouChina
| | - Haolan Gong
- Department of HematologyThe First Affiliated Hospital of Yangtze UniversityJingzhouChina
| | - Xiaoru Gao
- Department of HematologyThe First Affiliated Hospital of Yangtze UniversityJingzhouChina
| | - Xin Liu
- Department of HematologyThe First Affiliated Hospital of Yangtze UniversityJingzhouChina
| | - Yixin Gong
- Department of HematologyThe First Affiliated Hospital of Yangtze UniversityJingzhouChina
| | - Bo Luo
- Department of Radiation OncologyHubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Yanhua Zhang
- Department of HematologyThe First Affiliated Hospital of Yangtze UniversityJingzhouChina
- Department of PathophysiologySchool of Basic Medicine, Health Science Center, Yangtze UniversityJingzhouChina
| | - Xiaochun Peng
- Department of HematologyThe First Affiliated Hospital of Yangtze UniversityJingzhouChina
- Department of PathophysiologySchool of Basic Medicine, Health Science Center, Yangtze UniversityJingzhouChina
| | - Gai Liang
- Department of Radiation OncologyHubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| |
Collapse
|
37
|
Weyand CM, Goronzy JJ. Metabolic checkpoints in rheumatoid arthritis. Semin Arthritis Rheum 2025; 70S:152586. [PMID: 39550308 PMCID: PMC11761375 DOI: 10.1016/j.semarthrit.2024.152586] [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: 09/29/2024] [Accepted: 10/28/2024] [Indexed: 11/18/2024]
Abstract
BACKGROUND Rheumatoid Arthritis is a systemic autoimmune disease affecting 0.5-1 % of the population. Despite a growing therapeutic armamentarium, RA remains incurable, and many patients suffer significant morbidity over time. The strongest genetic risk derives from HLA class II polymorphisms, implicating T cells as pathogenic drivers. Innate immune cells, e.g. monocytes and macrophages (Mⱷ) contribute to chronic tissue inflammation through an array of pro-inflammatory functions but also present antigen to autoreactive T cells. Differentiation, survival, and effector functions of both T cells and Mⱷ are ultimately controlled by their bioenergetic and biosynthetic programs, identifying cellular metabolism as a critical disease mechanism in RA. OBJECTIVES Summarize current knowledge about metabolic conditions in the RA joint and disease-relevant metabolic circuits shaping the effector repertoire of RA T cells and Mⱷ. RESULTS The rheumatoid joint is a glucose deplete tissue environment, selecting for invading immune cells that can survive on non-glucose fuel sources. Inflamed synovium instead offers the amino acid glutamine and RA CD4+ T cells and RA Mⱷ rely on glutamine and glutamate to support their pathogenic functions. The metabolic hallmark of RA T cells is their low mitochondrial performance, resulting in low ATP production, low generation of reactive oxygen species (ROS) and low availability of tricarboxylic acid (TCA) cycle intermediates, all shifting RA T cells towards autoreactivity. The underlying defect stems from insufficient repair of mitochondrial DNA (mtDNA). Functional consequences include reversal of the TCA cycle, accumulation of citrate and lack of malate production. Excessive citrate promotes cytoskeletal hyperacetylation, creating hypermigratory and tissue-invasive T cells. Surplus acetyl-CoA supports lipid droplet formation and lipotoxicity. Lack of malate production disrupts the malate-aspartate shuttle, restricts recovery of cytosolic NAD and drives the endoplasmic reticulum (ER) into expansion. The bioenergetically stressed ER accumulates TNF mRNA and turns RA T cells into TNF superproducers. ATP low production renders RA T cells susceptible to cell death, depositing highly inflammatory mtDNA in the tissue. Mitochondrial deficiency leads to a slowdown in glycolysis and pyruvate processing, such that RA CD4+ T cells shunt glucose towards the pentose phosphate pathway to support nucleotide synthesis and clonal proliferation. Metabolically deprived CD4+ T cells partner with Mⱷ that have highly functional mitochondria. A hallmark of RA Mⱷ is the high expression of the DNA binding protein RFX5, which co-ordinates adaptations to metabolic needs with function. RFX5 upregulates HLA-DR expression and induces the glutaminolytic enzyme glutamate dehydrogenase 1 (GLUD1), providing bioenergetic resources for antigen presentation and survival in the tissue. In essence, RA CD4+ T cells and Mⱷ function in a metabolically challenging environment and rewire their cellular metabolism to survive. Metabolic adaptations promote immunostimulation and tissue inflammation, triggering and sustaining rheumatoid synovitis.
Collapse
Affiliation(s)
- Cornelia M Weyand
- Department of Medicine, Mayo Clinic Alix School of Medicine, Rochester, MN, 55905, USA; Department of Immunology, Mayo Clinic College of Medicine and Science, Rochester, MN, 55905, USA; Department of Cardiovascular Medicine, Mayo Clinic Alix School of Medicine, Rochester, MN, USA; Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| | - Jörg J Goronzy
- Department of Medicine, Mayo Clinic Alix School of Medicine, Rochester, MN, 55905, USA; Department of Immunology, Mayo Clinic College of Medicine and Science, Rochester, MN, 55905, USA; Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| |
Collapse
|
38
|
Wang W, Wu B, Hao M, Chen S, Cong R, Wu W, Wang P, Zhang Q, Jia P, Song Y, Liu B, Qu S, Pei JF, Li D, Zhang N. Positive feedback loop involving AMPK and CLYBL acetylation links metabolic rewiring and inflammatory responses. Cell Death Dis 2025; 16:41. [PMID: 39863605 PMCID: PMC11762313 DOI: 10.1038/s41419-025-07362-0] [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: 08/23/2024] [Revised: 12/08/2024] [Accepted: 01/16/2025] [Indexed: 01/27/2025]
Abstract
Metabolic rewiring underlies effective macrophages defense to respond disease microenvironment. However, the underlying mechanisms driving metabolic rewiring to enhance macrophage effector functions remain unclear. Here, we demonstrated that the metabolic reprogramming in inflammatory macrophages depended on the acetylation of CLYBL, a citramalyl-CoA lyase, at lysine 154 (K154), and blocking CLYBL-K154 acetylation restricted the release of pro-inflammatory factors. Mechanistically, we found a crucial AMPK-CLYBL acetylation positive feedback loop, triggered by toll-like receptors (TLRs), involving AMPK hypophosphorylation and CLYBL hyperacetylation. The deacetylase enzyme SIRT2 acted as the bridge between AMPK phosphorylation and CLYBL acetylation, thereby regulating macrophage polarization and the release of pro-inflammatory cytokines. Furthermore, CLYBL hypoacetylation decreased monocyte infiltration, thereby alleviating cardiac remodeling. These findings suggest that the AMPK-CLYBL acetylation positive feedback loop serves as a metabolic switch driving inflammatory response and inhibiting CLYBL-K154 acetylation may offer a promising therapeutic strategy for inflammatory response-related disorders.
Collapse
Affiliation(s)
- Wenke Wang
- Center of Reproductive Medicine, Shengjing Hospital of China Medical University, Shenyang, 110004, China
- NHC Key Laboratory of Advanced Reproductive Medicine and Fertility (China Medical University), National Health Commission, Shenyang, 110004, China
| | - Boquan Wu
- Department of Cardiology, The First Hospital of China Medical University, Shenyang, Liaoning, 110001, China
| | - Mingjun Hao
- Center of Reproductive Medicine, Shengjing Hospital of China Medical University, Shenyang, 110004, China
- NHC Key Laboratory of Advanced Reproductive Medicine and Fertility (China Medical University), National Health Commission, Shenyang, 110004, China
| | - Sichong Chen
- Department of Cardiology, The First Hospital of China Medical University, Shenyang, Liaoning, 110001, China
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Ruiting Cong
- Center of Reproductive Medicine, Shengjing Hospital of China Medical University, Shenyang, 110004, China
- NHC Key Laboratory of Advanced Reproductive Medicine and Fertility (China Medical University), National Health Commission, Shenyang, 110004, China
| | - Wenjie Wu
- Center of Reproductive Medicine, Shengjing Hospital of China Medical University, Shenyang, 110004, China
- NHC Key Laboratory of Advanced Reproductive Medicine and Fertility (China Medical University), National Health Commission, Shenyang, 110004, China
| | - Pengbo Wang
- Department of Cardiology, The First Hospital of China Medical University, Shenyang, Liaoning, 110001, China
| | - Qiaoyi Zhang
- Department of Cardiology, The First Hospital of China Medical University, Shenyang, Liaoning, 110001, China
- China Medical University School of Public Health, Shenyang, 110122, China
| | - Pengyu Jia
- Department of Cardiology, The First Hospital of China Medical University, Shenyang, Liaoning, 110001, China
| | - Yuequn Song
- Department of Neurosurgery, the Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, 110032, China
| | - Bo Liu
- Department of Cardiac Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, 110001, China.
| | - Siyao Qu
- Department of Medical Genetics, China Medical University, Shenyang, Liaoning, 110122, China.
| | - Jian-Fei Pei
- Department of Medical Genetics, China Medical University, Shenyang, Liaoning, 110122, China.
| | - Da Li
- Center of Reproductive Medicine, Shengjing Hospital of China Medical University, Shenyang, 110004, China.
- NHC Key Laboratory of Advanced Reproductive Medicine and Fertility (China Medical University), National Health Commission, Shenyang, 110004, China.
| | - Naijin Zhang
- NHC Key Laboratory of Advanced Reproductive Medicine and Fertility (China Medical University), National Health Commission, Shenyang, 110004, China.
- Department of Cardiology, The First Hospital of China Medical University, Shenyang, Liaoning, 110001, China.
- Institute of Health Sciences, China Medical University, Shenyang, Liaoning, 110122, China.
| |
Collapse
|
39
|
Zhang X, Wen J, Pan Z, Liu Y, Zhu Y. Celastrus orbiculatus Thunb. extract inhibits inflammatory metabolic adaptation in macrophages and regulates polarization via modulating PKM2. Int Immunopharmacol 2025; 144:113665. [PMID: 39591823 DOI: 10.1016/j.intimp.2024.113665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 11/05/2024] [Accepted: 11/15/2024] [Indexed: 11/28/2024]
Abstract
Precancerous lesions of gastric cancer (PLGC) are considered critical stages for the prevention and treatment of gastric cancer (GC), with gastric mucosal inflammation being a prerequisite for PLGC. Macrophages, integral to the immune system, typically respond to external stimuli triggering inflammation. Celastrus orbiculatus Thunb. extract (COE) has been shown to exhibit anti-inflammatory effects in treating PLGC. However, it remains unclear how COE modulates macrophage metabolic adaptation and polarization in the inflammatory response to reverse PLGC. This study utilized a composite modeling approach to establish a PLGC mouse model, assessing COE's impact on polarization and metabolic adaptation markers such as inflammatory factors in gastric mucosa and RAW264.7 macrophages. The results confirm that COE significantly reduces M1 macrophage polarization markers while increasing M2 macrophage polarization markers and lowering inflammatory factor levels. Additionally, COE effectively inhibits the expression of pyruvate kinase M2 (PKM2). Our findings suggest that COE may act through regulating PKM2 expression to modulate inflammatory responses and reverse PLGC.
Collapse
Affiliation(s)
- Xiaoze Zhang
- Chinese Integrative Medicine Oncology Depatrment, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230000, China
| | - Junsong Wen
- Chinese Integrative Medicine Oncology Depatrment, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230000, China
| | - Ziwei Pan
- Chinese Integrative Medicine Oncology Depatrment, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230000, China
| | - Yanqing Liu
- Institute of Traditional Chinese Medicine & Western Medicine, School of Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, China
| | - Yaodong Zhu
- Chinese Integrative Medicine Oncology Depatrment, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230000, China.
| |
Collapse
|
40
|
Babaniamansour P, Jacho D, Rabino A, Garcia-Mata R, Yildirim-Ayan E. Synergetic role of TRPV4 inhibitor and mechanical loading on reducing inflammation. Front Immunol 2025; 15:1456042. [PMID: 39850885 PMCID: PMC11756524 DOI: 10.3389/fimmu.2024.1456042] [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: 06/27/2024] [Accepted: 12/04/2024] [Indexed: 01/25/2025] Open
Abstract
Resolution of inflammation is essential for normal tissue healing and regeneration, with macrophages playing a key role in regulating this process through phenotypic changes from a pro-inflammatory to an anti-inflammatory state. Pharmacological and mechanical (mechanotherapy) techniques can be employed to polarize macrophages toward an anti-inflammatory phenotype, thereby diminishing inflammation. One clinically relevant pharmacological approach is the inhibition of Transient Receptor Potential Vanilloid 4 (TRPV4). This study investigates the effects of various mechanical loading amplitudes (0%, 3%, and 6%) and TRPV4 inhibition (10 µM RN-1734) on the phenotypic commitments of pro-inflammatory (M1) macrophages within three-dimensional (3D) collagen matrices. M1 macrophages exposed to 3% mechanical strain exhibited upregulated pro-inflammatory responses, including increased pro-inflammatory gene expression and enhanced proteolytic activity within the extracellular matrix. TRPV4 inhibition partially mitigated this inflammation. Notably, 6% mechanical strain combined with TRPV4 inhibition suppressed Mitogen-Activated Protein Kinase (MAPK) expression, leading to reduced pro-inflammatory gene expression and increased anti-inflammatory markers such as CD206. Gene expression analysis further demonstrated significant reductions in pro-inflammatory gene expression and a synergistic promotion of anti-inflammatory phenotypes under TRPV4 inhibition at 6% mechanical strain. Surface protein analysis via immunohistochemistry confirmed these phenotypic shifts, highlighting changes in the expression of CD80 (pro-inflammatory) and CD206 (anti-inflammatory) markers, alongside F-actin and nuclear staining. This research suggests that TRPV4 inhibition, combined with specific mechanical loading (6%), can drive macrophages toward an anti-inflammatory state, thereby may promote inflammation resolution and tissue repair.
Collapse
Affiliation(s)
- Parto Babaniamansour
- Department of Bioengineering, College of Engineering, University of Toledo, Toledo, OH, United States
| | - Diego Jacho
- Department of Bioengineering, College of Engineering, University of Toledo, Toledo, OH, United States
| | - Agustin Rabino
- Department of Biological Sciences, University of Toledo, Toledo, OH, United States
| | - Rafael Garcia-Mata
- Department of Biological Sciences, University of Toledo, Toledo, OH, United States
| | - Eda Yildirim-Ayan
- Department of Bioengineering, College of Engineering, University of Toledo, Toledo, OH, United States
| |
Collapse
|
41
|
Zhang P, Watari K, Karin M. Innate immune cells link dietary cues to normal and abnormal metabolic regulation. Nat Immunol 2025; 26:29-41. [PMID: 39747429 PMCID: PMC12040443 DOI: 10.1038/s41590-024-02037-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 10/24/2024] [Indexed: 01/04/2025]
Abstract
A slew of common metabolic disorders, including type 2 diabetes, metabolic dysfunction-associated steatotic liver disease and steatohepatitis, are exponentially increasing in our sedentary and overfed society. While macronutrients directly impact metabolism and bioenergetics, new evidence implicates immune cells as critical sensors of nutritional cues and important regulators of metabolic homeostasis. A deeper interrogation of the intricate and multipartite interactions between dietary components, immune cells and metabolically active tissues is needed for a better understanding of metabolic regulation and development of new treatments for common metabolic diseases. Responding to macronutrients and micronutrients, immune cells play pivotal roles in interorgan communication between the microbiota, small intestine, metabolically active cells including hepatocytes and adipocytes, and the brain, which controls feeding behavior and energy expenditure. This Review focuses on the response of myeloid cells and innate lymphocytes to dietary cues, their cross-regulatory interactions and roles in normal and aberrant metabolic control.
Collapse
Affiliation(s)
- Peng Zhang
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Kosuke Watari
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Michael Karin
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, School of Medicine, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
42
|
Han X, Wu X, Liu F, Chen H, Hou H. Inhibition of LPS-induced inflammatory response in RAW264.7 cells by natural Chlorogenic acid isomers involved with AKR1B1 inhibition. Bioorg Med Chem 2024; 114:117942. [PMID: 39396466 DOI: 10.1016/j.bmc.2024.117942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 10/05/2024] [Accepted: 10/06/2024] [Indexed: 10/15/2024]
Abstract
Inflammation is the physiological response of the immune system to injury or infection, typically manifested by local tissue congestion, swelling, heat, and pain. Prolonged or excessive inflammation can lead to tissue damage and the development of many diseases. The anti-inflammatory effects of natural ingredients have been extensively researched and confirmed. This study investigated the effects of Chlorogenic acid (CGA) isomers -- 3-Caffeolyquninic acid (3-CQA), 4-Caffeolyquninic acid (4-CQA), and 5-Caffeolyquninic acid (5-CQA) -- on the inflammatory response and oxidative stress reaction induced by LPS in RAW264.7 cells. Overall, 3-CQA exhibited the most significant reduction in levels of TNF-α, IL-6, NO, and ROS. 4-CQA showed superior inhibition of TNF-α compared to 5-CQA (p < 0.05), while no significant difference in other parameters. We further used DARTS and CETSA to demonstrate that CGA isomers have stable affinity with AKR1B1. As a positive control, the AKR1B1 antagonist epalrestat exhibited similar effects to the CGA isomers. 3-CQA having the smallest half-inhibitory concentration (IC50) for AKR1B1, while 4-CQA and 5-CQA have similar values. AutoDock simulations of the docking conformations revealed minimal differences in the average binding energies of the CGA isomers. The main differences were that VAL47 formed a hydrogen bond with 3-CQA, whereas GLN49 formed hydrogen bonds with 4-CQA and 5-CQA. Additionally, the number of hydrophobic bonds involving PHE122 and LEU300 varies. Our conclusion is that differences in non-covalent interactions result in the varying inhibitory abilities of CGA isomers on AKR1B1, which further affect the anti-inflammatory and antioxidant effects of CGA isomers.
Collapse
Affiliation(s)
- Xu Han
- Anhui Institute of Optics and Fine Mechanics, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, PR China; University of Science and Technology of China, Hefei 230026, PR China; China National Tobacco Quality Supervision & Test Center, Zhengzhou 450001, PR China; Key Laboratory of Tobacco Biological Effects, Zhengzhou, PR China
| | - Xiaqing Wu
- China National Tobacco Quality Supervision & Test Center, Zhengzhou 450001, PR China; Key Laboratory of Tobacco Biological Effects, Zhengzhou, PR China
| | - Fanglin Liu
- Anhui Institute of Optics and Fine Mechanics, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, PR China; University of Science and Technology of China, Hefei 230026, PR China.
| | - Huan Chen
- China National Tobacco Quality Supervision & Test Center, Zhengzhou 450001, PR China; Key Laboratory of Tobacco Biological Effects, Zhengzhou, PR China.
| | - Hongwei Hou
- China National Tobacco Quality Supervision & Test Center, Zhengzhou 450001, PR China; Key Laboratory of Tobacco Biological Effects, Zhengzhou, PR China.
| |
Collapse
|
43
|
Zhang T, Lu Z, Liu J, Tao Y, Si Y, Ye J, Cao S, Zhu B. Host Innate and Adaptive Immunity Against African Swine Fever Virus Infection. Vaccines (Basel) 2024; 12:1278. [PMID: 39591181 PMCID: PMC11599025 DOI: 10.3390/vaccines12111278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 11/09/2024] [Accepted: 11/12/2024] [Indexed: 11/28/2024] Open
Abstract
Africa swine fever virus (ASFV) is the causative agent of African swine fever (ASF), a highly contagious hemorrhagic disease that can result in up to 100% lethality in both wild and domestic swine, regardless of breed or age. The ongoing ASF pandemic poses significant threats to the pork industry and food security, with serious implications for the sanitary and socioeconomic system. Due to the limited understanding of ASFV pathogenesis and immune protection mechanisms, there are currently no safe and effective vaccines or specific treatments available, complicating efforts for prevention and control. This review summarizes the current understanding of the intricate interplay between ASFV and the host immune system, encompassing both innate and adaptive immune responses to ASFV infection, as well as insights into ASFV pathogenesis and immunosuppression. We aim to provide comprehensive information to support fundamental research on ASFV, highlighting existing gaps and suggesting future research directions. This work may serve as a theoretical foundation for the rational design of protective vaccines against this devastating viral disease.
Collapse
Affiliation(s)
- Tianqi Zhang
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (T.Z.); (Z.L.); (J.L.); (Y.T.); (Y.S.); (J.Y.); (S.C.)
- Frontiers Science Center for Animal Breeding and Sustainable Production, Huazhong Agricultural University, Wuhan 430070, China
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan 430070, China
| | - Zixun Lu
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (T.Z.); (Z.L.); (J.L.); (Y.T.); (Y.S.); (J.Y.); (S.C.)
- Frontiers Science Center for Animal Breeding and Sustainable Production, Huazhong Agricultural University, Wuhan 430070, China
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan 430070, China
| | - Jia Liu
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (T.Z.); (Z.L.); (J.L.); (Y.T.); (Y.S.); (J.Y.); (S.C.)
- Frontiers Science Center for Animal Breeding and Sustainable Production, Huazhong Agricultural University, Wuhan 430070, China
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan 430070, China
| | - Yang Tao
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (T.Z.); (Z.L.); (J.L.); (Y.T.); (Y.S.); (J.Y.); (S.C.)
- Frontiers Science Center for Animal Breeding and Sustainable Production, Huazhong Agricultural University, Wuhan 430070, China
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan 430070, China
| | - Youhui Si
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (T.Z.); (Z.L.); (J.L.); (Y.T.); (Y.S.); (J.Y.); (S.C.)
- Frontiers Science Center for Animal Breeding and Sustainable Production, Huazhong Agricultural University, Wuhan 430070, China
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan 430070, China
| | - Jing Ye
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (T.Z.); (Z.L.); (J.L.); (Y.T.); (Y.S.); (J.Y.); (S.C.)
- Frontiers Science Center for Animal Breeding and Sustainable Production, Huazhong Agricultural University, Wuhan 430070, China
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan 430070, China
| | - Shengbo Cao
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (T.Z.); (Z.L.); (J.L.); (Y.T.); (Y.S.); (J.Y.); (S.C.)
- Frontiers Science Center for Animal Breeding and Sustainable Production, Huazhong Agricultural University, Wuhan 430070, China
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan 430070, China
| | - Bibo Zhu
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (T.Z.); (Z.L.); (J.L.); (Y.T.); (Y.S.); (J.Y.); (S.C.)
- Frontiers Science Center for Animal Breeding and Sustainable Production, Huazhong Agricultural University, Wuhan 430070, China
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan 430070, China
| |
Collapse
|
44
|
Sheu KM, Pimplaskar A, Hoffmann A. Single-cell stimulus-response gene expression trajectories reveal the stimulus specificities of dynamic responses by single macrophages. Mol Cell 2024; 84:4095-4110.e6. [PMID: 39413794 PMCID: PMC11560543 DOI: 10.1016/j.molcel.2024.09.023] [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: 11/03/2023] [Revised: 07/05/2024] [Accepted: 09/19/2024] [Indexed: 10/18/2024]
Abstract
Macrophages induce the expression of hundreds of genes in response to immune threats. However, current technology limits our ability to capture single-cell inducible gene expression dynamics. Here, we generated high-resolution time series single-cell RNA sequencing (scRNA-seq) data from mouse macrophages responding to six stimuli, and imputed ensembles of real-time single-cell gene expression trajectories (scGETs). We found that dynamic information contained in scGETs substantially contributes to macrophage stimulus-response specificity (SRS). Dynamic information also identified correlations between immune response genes, indicating biological coordination. Furthermore, we showed that the microenvironmental context of polarizing cytokines profoundly affects scGETs, such that measuring response dynamics offered clearer discrimination of the polarization state of individual macrophage cells than single time-point measurements. Our findings highlight the important contribution of dynamic information contained in the single-cell expression responses of immune genes in characterizing the SRS and functional states of macrophages.
Collapse
Affiliation(s)
- Katherine M Sheu
- Department of Microbiology, Immunology, and Molecular Genetics, and Institute for Quantitative and Computational Biosciences, University of California, Los Angeles, 611 Charles E. Young Dr S, Los Angeles, CA 90093, USA
| | - Aditya Pimplaskar
- Department of Microbiology, Immunology, and Molecular Genetics, and Institute for Quantitative and Computational Biosciences, University of California, Los Angeles, 611 Charles E. Young Dr S, Los Angeles, CA 90093, USA
| | - Alexander Hoffmann
- Department of Microbiology, Immunology, and Molecular Genetics, and Institute for Quantitative and Computational Biosciences, University of California, Los Angeles, 611 Charles E. Young Dr S, Los Angeles, CA 90093, USA.
| |
Collapse
|
45
|
Gaigeard N, Cardon A, Le Goff B, Guicheux J, Boutet MA. Unveiling the macrophage dynamics in osteoarthritic joints: From inflammation to therapeutic strategies. Drug Discov Today 2024; 29:104187. [PMID: 39306233 DOI: 10.1016/j.drudis.2024.104187] [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: 06/19/2024] [Revised: 09/06/2024] [Accepted: 09/17/2024] [Indexed: 09/29/2024]
Abstract
Osteoarthritis (OA) is an incurable, painful, and debilitating joint disease affecting over 500 million people worldwide. The OA joint tissues are infiltrated by various immune cells, particularly macrophages, which are able to induce or perpetuate inflammation. Notably, synovitis and its macrophage component represent a target of interest for developing treatments. In this review, we describe the latest advances in understanding the heterogeneity of macrophage origins, phenotypes, and functions in the OA joint and the effect of current symptomatic therapies on these cells. We then highlight the therapeutic potential of anticytokines/chemokines, nano- and microdrug delivery, and future strategies to modulate macrophage functions in OA.
Collapse
Affiliation(s)
- Nicolas Gaigeard
- Nantes Université, Oniris, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR1229, F-44000 Nantes, France
| | - Anaïs Cardon
- Nantes Université, Oniris, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR1229, F-44000 Nantes, France
| | - Benoit Le Goff
- Nantes Université, Oniris, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR1229, F-44000 Nantes, France
| | - Jérôme Guicheux
- Nantes Université, Oniris, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR1229, F-44000 Nantes, France
| | - Marie-Astrid Boutet
- Nantes Université, Oniris, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR1229, F-44000 Nantes, France; Centre for Experimental Medicine & Rheumatology, William Harvey Research Institute and Barts and The London School of Medicine and Dentistry, Queen Mary University of London, EC1M6BQ London, UK.
| |
Collapse
|
46
|
Song G, Li M, Zhou B, Qi H, Guo J. Streptococcus mutans outer membrane vesicles affect inflammasome activation and the glycolysis of macrophages. Microb Pathog 2024; 196:106994. [PMID: 39366588 DOI: 10.1016/j.micpath.2024.106994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 09/21/2024] [Accepted: 10/01/2024] [Indexed: 10/06/2024]
Abstract
Recent studies indicate that bacterial outer membrane vesicles (OMVs) play a significant role in bacterial virulence and pathogenicity. Streptococcus mutans (S. mutans), a principal pathogen in dental caries, secretes a substantial number of OMVs. However, the impact of S. mutans OMVs on oral health and their underlying pathogenic mechanisms remain poorly understood. Macrophages were the initial innate immune cells to respond to bacterial invaders and their products. Therefore, we purified S. mutans OMVs, which stimulated macrophages. Compared to controls, RT-PCR and ELISA analyses revealed that S. mutans OMVs significantly increased the production of IL-1β, IL-6, TNF-α and IL-8, with IL-1β being notably elevated. IL-1β production and secretion are tightly regulated by the inflammasome. Western blot analyses demonstrated that S. mutans OMVs upregulated the expression of inflammasome components, including NLRP3, NLRC4, ASC and AIM2, with a marked increase in NLRP3 expression. Silencing different inflammasome components with siRNA revealed a reduction in IL-1β secretion induced by S. mutans OMVs, particularly through NLRP3. Additionally, ATP production and K+ efflux were found to be crucial for NLRP3 activation. Prolonged stimulation with S. mutans OMVs resulted in increased lactate production and elevated expression of glycolysis-related genes Glut-1, PFKFB3, and HK I, indicating that S. mutans OMVs significantly induce macrophage glycolysis. Furthermore, S. mutans OMVs were shown to enhance biofilm formation, increase S. mutans colonisation on epithelial cells, and inhibit macrophage phagocytosis, thereby improving the survival of S. mutans in the oral cavity. In summary, S. mutans OMVs promote the survival of S. mutans in the mouth through multiple mechanisms, potentially influencing the development of dental caries.
Collapse
Affiliation(s)
- Gongyuan Song
- The Second Hospital of Shijiazhuang, Shijiazhuang, 050000, China
| | - Min Li
- Handan Stomatology Hospital, Handan, 056000, China
| | - Bing Zhou
- Cangzhou People's Hospital, Cangzhou, 061000, China
| | - Hongguang Qi
- Gucheng County Hospital of Hebei Provence, Gucheng, 253800, China
| | - Jie Guo
- Department of Stomatology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050000, China.
| |
Collapse
|
47
|
Chen H, Song J, Zeng L, Zha J, Zhu J, Chen A, Liu Y, Dong Z, Chen G. Dietary sodium modulates mTORC1-dependent trained immunity in macrophages to accelerate CKD development. Biochem Pharmacol 2024; 229:116505. [PMID: 39181336 DOI: 10.1016/j.bcp.2024.116505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 08/15/2024] [Accepted: 08/20/2024] [Indexed: 08/27/2024]
Abstract
Chronic Kidney Disease (CKD) is a significant global health issue linked to dietary habits, especially high salt intake. However, the precise mechanisms driving this progression remain incompletely understood. This study reveals that a high-salt diet intensifies macrophage trained immunity, leading to a marked pro-inflammatory response upon repeated pathogenic exposures, as evidenced by increased renal damage and fibrosis. Under high-salt conditions, there was an induction of CD45+F4/80+ macrophage infiltration into the renal tissue, accompanied by heightened production of inflammatory cytokines. Distinct responses were observed between circulating and resident renal macrophages to a high-salt diet, with a notable upsurge in the migration of pro-inflammatory macrophages, driven by CCL2-CCR2 signaling and aberrant mTORC1 pathway activation. Treatment with rapamycin-liposome effectively reduced this inflammatory cascade by mitigating mTORC1 signaling. Transplantation of monocytes from CKD mice with a high-salt diet significantly exacerbates renal inflammatory damage in the host mice, showing increased migratory tendency and inflammatory activity. The cell co-culture experiment further confirmed that macrophages derived from CKD mice, particularly those under conditions of high salt exposure, significantly induced apoptosis and inflammatory responses in renal tubular cells. Taken together, recurrent exposure to LPS elicits the activation of trained immunity, consequently augmenting inflammatory response of monocytes/macrophages in the involved kidneys. The high-salt diet exacerbates this phenomenon, attributable at least in part to the overactivation of the mTORC1 pathway. This research emphasizes the importance of dietary modulation and targeted immunological interventions in slowing CKD progression, providing new insights into mTORC1-mediated pathophysiological mechanisms and potential management strategies for CKD.
Collapse
Affiliation(s)
- Huihui Chen
- Department of Ophthalmology, the Second Xiangya Hospital of Central South University, Changsha, China
| | - Jie Song
- Department of Nephrology, the Second Xiangya Hospital of Central South University, Changsha, China
| | - Li Zeng
- Department of Nephrology, the Second Xiangya Hospital of Central South University, Changsha, China
| | - Jie Zha
- Hunan Key Laboratory of Kidney Disease and Blood Purification, the Second Xiangya Hospital of Central South University, Changsha, China
| | - Jiefu Zhu
- Department of Transplantation, Renmin Hospital of Wuhan University, Wuhan, China
| | - Anqun Chen
- Department of Nephrology, the Second Xiangya Hospital of Central South University, Changsha, China; Hunan Key Laboratory of Kidney Disease and Blood Purification, the Second Xiangya Hospital of Central South University, Changsha, China
| | - Yu Liu
- Department of Nephrology, the Second Xiangya Hospital of Central South University, Changsha, China; Hunan Key Laboratory of Kidney Disease and Blood Purification, the Second Xiangya Hospital of Central South University, Changsha, China
| | - Zheng Dong
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University and Charlie Norwood Veterans Affairs Medical Center, Augusta, GA, USA
| | - Guochun Chen
- Department of Nephrology, the Second Xiangya Hospital of Central South University, Changsha, China; Hunan Key Laboratory of Kidney Disease and Blood Purification, the Second Xiangya Hospital of Central South University, Changsha, China.
| |
Collapse
|
48
|
Xu R, Vujić N, Bianco V, Reinisch I, Kratky D, Krstic J, Prokesch A. Lipid-associated macrophages between aggravation and alleviation of metabolic diseases. Trends Endocrinol Metab 2024; 35:981-995. [PMID: 38705759 DOI: 10.1016/j.tem.2024.04.009] [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: 01/20/2024] [Revised: 04/09/2024] [Accepted: 04/10/2024] [Indexed: 05/07/2024]
Abstract
Lipid-associated macrophages (LAMs) are phagocytic cells with lipid-handling capacity identified in various metabolic derangements. During disease development, they locate to atherosclerotic plaques, adipose tissue (AT) of individuals with obesity, liver lesions in steatosis and steatohepatitis, and the intestinal lamina propria. LAMs can also emerge in the metabolically demanding microenvironment of certain tumors. In this review, we discuss major questions regarding LAM recruitment, differentiation, and self-renewal, and, ultimately, their acute and chronic functional impact on the development of metabolic diseases. Further studies need to clarify whether and under which circumstances LAMs drive disease progression or resolution and how their phenotype can be modulated to ameliorate metabolic disorders.
Collapse
Affiliation(s)
- Ruonan Xu
- Gottfried Schatz Research Center for Cell Signaling, Metabolism, and Aging, Division of Cell Biology, Histology, and Embryology, Medical University of Graz, Graz, Austria
| | - Nemanja Vujić
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Valentina Bianco
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Isabel Reinisch
- Institute of Food Nutrition and Health, Department of Health Sciences and Technology, Eidgenössische Technische Hochschule Zürich (ETH), Schwerzenbach, Switzerland
| | - Dagmar Kratky
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria; BioTechMed-Graz, Graz, Austria
| | - Jelena Krstic
- Gottfried Schatz Research Center for Cell Signaling, Metabolism, and Aging, Division of Cell Biology, Histology, and Embryology, Medical University of Graz, Graz, Austria; BioTechMed-Graz, Graz, Austria
| | - Andreas Prokesch
- Gottfried Schatz Research Center for Cell Signaling, Metabolism, and Aging, Division of Cell Biology, Histology, and Embryology, Medical University of Graz, Graz, Austria; BioTechMed-Graz, Graz, Austria.
| |
Collapse
|
49
|
Xiao J, Deng Y, Xie J, Liu H, Yang Q, Zhang Y, Huang X, Cao Z. Apoptotic vesicles from macrophages exacerbate periodontal bone resorption in periodontitis via delivering miR-143-3p targeting Igfbp5. J Nanobiotechnology 2024; 22:658. [PMID: 39456001 PMCID: PMC11515254 DOI: 10.1186/s12951-024-02934-2] [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: 07/24/2024] [Accepted: 10/14/2024] [Indexed: 10/28/2024] Open
Abstract
ABSTRCT BACKGROUND: Apoptotic vesicles (ApoVs), which are extracellular vesicles released by apoptotic cells, have been reported to exhibit substantial therapeutic potential for inflammatory diseases and tissue regeneration. While extensive research has been dedicated to mesenchymal stem cells (MSCs), the investigation into immune cell-derived ApoVs remains limited, particularly regarding the function and fate of macrophage-derived ApoVs in the context of periodontitis (PD). RESULTS Our study corroborates the occurrence and contribution of resident macrophage apoptosis in Porphyromonas gingivalis (Pg)-associated PD. The findings unveil the pivotal role played by apoptotic macrophages and their derived ApoVs in orchestrating periodontal bone remodeling. The enrichments of diverse functional miRNAs within these ApoVs are discerned through sequencing techniques. Moreover, our study elucidates that the macrophage-derived ApoVs predominantly deliver miR-143-3p, targeting insulin-like growth factor-binding protein 5 (IGFBP5), thereby disrupting periodontal bone homeostasis. CONCLUSIONS Our study reveals that macrophages in Pg-associated PD undergo apoptosis and generate miR-143-3p-enriched ApoVs to silence IGFBP5, resulting in the perturbation of osteogenic-osteoclastic balance and the ensuing periodontal bone destruction. Accordingly, interventions targeting miR-143-3p in macrophages or employment of apoptosis inhibitor Z-VAD hold promise as effective therapeutic strategies for the management of PD.
Collapse
Affiliation(s)
- Junhong Xiao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
- Qingdao Stomatological Hospital Affiliated to Qingdao University, No.17 Dexian Road, Shinan District, Qingdao, 266001, Shandong Province, China
| | - Yifei Deng
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Jirong Xie
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Heyu Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Qiudong Yang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Yufeng Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China.
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, 430071, China.
- Department of Oral Implantology, School & Hospital of Stomatology, Wuhan University, 237 Luoyu Road, Hongshan District, Wuhan, 430079, China.
| | - Xin Huang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China.
- Department of Periodontology, School & Hospital of Stomatology, Wuhan University, 237 Luoyu Road, Hongshan District, Wuhan, 430079, China.
| | - Zhengguo Cao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China.
- Department of Periodontology, School & Hospital of Stomatology, Wuhan University, 237 Luoyu Road, Hongshan District, Wuhan, 430079, China.
| |
Collapse
|
50
|
Cao C, Liu W, Guo X, Weng S, Chen Y, Luo Y, Wang S, Zhu B, Liu Y, Peng D. Identification and validation of efferocytosis-related biomarkers for the diagnosis of metabolic dysfunction-associated steatohepatitis based on bioinformatics analysis and machine learning. Front Immunol 2024; 15:1460431. [PMID: 39497821 PMCID: PMC11532026 DOI: 10.3389/fimmu.2024.1460431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Accepted: 10/02/2024] [Indexed: 11/07/2024] Open
Abstract
Background Metabolic dysfunction-associated steatohepatitis (MASH) is a highly prevalent liver disease globally, with a significant risk of progressing to cirrhosis and even liver cancer. Efferocytosis, a process implicated in a broad spectrum of chronic inflammatory disorders, has been reported to be associated with the pathogenesis of MASH; however, its precise role remains obscure. Thus, we aimed to identify and validate efferocytosis linked signatures for detection of MASH. Methods We retrieved gene expression patterns of MASH from the GEO database and then focused on assessing the differential expression of efferocytosis-related genes (EFRGs) between MASH and control groups. This analysis was followed by a series of in-depth investigations, including protein-protein interaction (PPI), correlation analysis, and functional enrichment analysis, to uncover the molecular interactions and pathways at play. To screen for biomarkers for diagnosis, we applied machine learning algorithm to identify hub genes and constructed a clinical predictive model. Additionally, we conducted immune infiltration and single-cell transcriptome analyses in both MASH and control samples, providing insights into the immune cell landscape and cellular heterogeneity in these conditions. Results This research pinpointed 39 genes exhibiting a robust correlation with efferocytosis in MASH. Among these, five potential diagnostic biomarkers-TREM2, TIMD4, STAB1, C1QC, and DYNLT1-were screened using two distinct machine learning models. Subsequent external validation and animal experimentation validated the upregulation of TREM2 and downregulation of TIMD4 in MASH samples. Notably, both TREM2 and TIMD4 demonstrated area under the curve (AUC) values exceeding 0.9, underscoring their significant potential in facilitating the diagnosis of MASH. Conclusion Our study comprehensively elucidated the relationship between MASH and efferocytosis, constructing a favorable diagnostic model. Furthermore, we identified potential therapeutic targets for MASH treatment and offered novel insights into unraveling the underlying mechanisms of this disease.
Collapse
Affiliation(s)
- Chenghui Cao
- Department of Cardiology, Research Institute of Blood Lipids and Atherosclerosis, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Wenwu Liu
- Department of Cardiology, Research Institute of Blood Lipids and Atherosclerosis, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xin Guo
- Department of Cardiology, Research Institute of Blood Lipids and Atherosclerosis, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Shuwei Weng
- Department of Cardiology, Research Institute of Blood Lipids and Atherosclerosis, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yang Chen
- Department of Cardiology, Research Institute of Blood Lipids and Atherosclerosis, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yonghong Luo
- Department of Cardiology, Research Institute of Blood Lipids and Atherosclerosis, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Shuai Wang
- Department of Cardiology, Research Institute of Blood Lipids and Atherosclerosis, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Botao Zhu
- Department of Cardiology, Research Institute of Blood Lipids and Atherosclerosis, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Yuxuan Liu
- Department of Cardiology, Research Institute of Blood Lipids and Atherosclerosis, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Daoquan Peng
- Department of Cardiology, Research Institute of Blood Lipids and Atherosclerosis, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|