1
|
Bhagwat A, Haldar T, Kanojiya P, Saroj SD. Bacterial metabolism in the host and its association with virulence. Virulence 2025; 16:2459336. [PMID: 39890585 PMCID: PMC11792850 DOI: 10.1080/21505594.2025.2459336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 01/13/2025] [Accepted: 01/22/2025] [Indexed: 02/03/2025] Open
Abstract
The host restricted pathogens are competently dependent on their respective host for nutritional requirements. The bacterial metabolic pathways are surprisingly varied and remarkably flexible that in turn help them to successfully overcome competition and colonise their host. The metabolic adaptation plays pivotal role in bacterial pathogenesis. The understanding of host-pathogen metabolic crosstalk needs to be prioritized to decipher host-pathogen interactions. The review focuses on various aspects of host pathogen interactions that majorly involves adaptation of bacterial metabolism to counteract immune mechanisms by rectifying metabolic cues that provides pathogen the idea of different anatomical sites and the local physiology of the host. The key set of metabolites that are recognized as centre of competition between host and its pathogens are also briefly discussed. The factors that control the timely expression of virulence of bacterial pathogens is poorly understood. The perspective presented herein will facilitate us with a broader view of molecular mechanisms that modulates the expression of virulence factors in bacterial pathogens. The knowledge of crosslinked metabolic pathways of bacteria and their host will serve to develop novel potential therapeutics.
Collapse
Affiliation(s)
- Amrita Bhagwat
- Symbiosis School of Biological Sciences, Symbiosis International (Deemed University), Pune, India
| | - Tiyasa Haldar
- Symbiosis School of Biological Sciences, Symbiosis International (Deemed University), Pune, India
| | - Poonam Kanojiya
- Symbiosis School of Biological Sciences, Symbiosis International (Deemed University), Pune, India
| | - Sunil D. Saroj
- Symbiosis School of Biological Sciences, Symbiosis International (Deemed University), Pune, India
| |
Collapse
|
2
|
Chen Z, Lin G, Ye K, Wang J, Tang M, Lai K, Yuan Y, Lin S, Dai X, Chen H, Ma H, Zhou J, Xu Y. Single-cell analysis of diquat-induced oxidative stress and its impact on organ-specific toxicity. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 297:118246. [PMID: 40327929 DOI: 10.1016/j.ecoenv.2025.118246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2025] [Revised: 04/13/2025] [Accepted: 04/23/2025] [Indexed: 05/08/2025]
Abstract
Diquat (DQ) is a commonly used herbicide, and its improper use can lead to multi-organ damage. The characteristics of multi-organ damage induced by DQ are not yet well understood. In this study, we conducted single-cell/single-nucleus RNA sequencing (scRNA-seq/snRNA-seq) on the lungs, liver, and kidneys of mice at consecutive time points (10 hours, 20 hours, and 36 hours) after DQ poisoning, as well as in a control group. In a multi-organ single-cell atlas comprising over 270,000 cells from mice, we found that DQ induces an oxidative stress microenvironment in endothelial and parenchymal cells, primarily characterized by activation of the oxidative phosphorylation pathway and an enhanced inflammatory response. This oxidative stress microenvironment prompted regulatory cell death in parenchymal and immune cells, releasing inflammatory factors and further exacerbating the oxidative environment. Additionally, metabolic reprogramming occurred in both parenchymal and immune cells under oxidative stress, leading to alterations in energy metabolism, reduced hepatic detoxification capabilities, and changes in the activation modes of immune cells, thereby intensifying tissue damage. Notably, no significant fibrosis was observed in the tissue damage caused by DQ, underscoring the importance of early intervention. Overall, using a mouse model, this study revealed the central role of the DQ-induced oxidative stress microenvironment in multi-organ damage and enhanced our understanding of the pathophysiology and complex molecular mechanisms underlying DQ-induced multi-organ injury.
Collapse
Affiliation(s)
- Zhimin Chen
- Department of Nephrology, Blood Purification Research Center, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China; Research Center for Metabolic Chronic Kidney Disease, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China; Department of Nephrology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Guo Lin
- Department of Intensive Care Unit, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
| | - Keng Ye
- Department of Nephrology, Blood Purification Research Center, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China; Research Center for Metabolic Chronic Kidney Disease, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China; Department of Nephrology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Junjie Wang
- Department of Nephrology, Blood Purification Research Center, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China; Research Center for Metabolic Chronic Kidney Disease, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China; Department of Nephrology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Min Tang
- Department of Nephrology, Blood Purification Research Center, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China; Research Center for Metabolic Chronic Kidney Disease, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China; Department of Nephrology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Kunmei Lai
- Department of Nephrology, Blood Purification Research Center, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China; Research Center for Metabolic Chronic Kidney Disease, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China; Department of Nephrology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Ying Yuan
- Department of Nephrology, Blood Purification Research Center, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China; Research Center for Metabolic Chronic Kidney Disease, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China; Department of Nephrology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Siyi Lin
- Department of Nephrology, Blood Purification Research Center, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China; Research Center for Metabolic Chronic Kidney Disease, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China; Department of Nephrology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Xingchen Dai
- Department of Nephrology, Blood Purification Research Center, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China; Research Center for Metabolic Chronic Kidney Disease, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China; Department of Nephrology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Hong Chen
- Department of Pathology, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
| | - Huabin Ma
- Central Laboratory, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
| | - Jianfu Zhou
- Central Laboratory, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
| | - Yanfang Xu
- Department of Nephrology, Blood Purification Research Center, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China; Research Center for Metabolic Chronic Kidney Disease, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China; Department of Nephrology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China; Central Laboratory, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China.
| |
Collapse
|
3
|
Molina-Lopez C, Hurtado-Navarro L, O'Neill LAJ, Pelegrin P. 4-octyl itaconate reduces human NLRP3 inflammasome constitutive activation with the cryopyrin-associated periodic syndrome p.R262W, p.D305N and p.T350M variants. Cell Mol Life Sci 2025; 82:209. [PMID: 40410596 PMCID: PMC12102053 DOI: 10.1007/s00018-025-05699-5] [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: 10/30/2024] [Revised: 03/27/2025] [Accepted: 04/06/2025] [Indexed: 05/25/2025]
Abstract
Cryopyrin-associated periodic syndrome (CAPS) is a condition characterized by dominant genetic variants in the NLRP3 gene, which lead to the formation of constitutively active inflammasomes. These inflammasomes play a crucial role in CAPS patients' inflammatory episodes, these being primarily driven by the production of interleukin (IL)-1b. Although treatment with IL-1 blockers is effective for CAPS, some patients develop refractory responses and adverse reactions to these therapies. Consequently, there is a need for novel treatments for CAPS patients. Promising candidates are the derivatives of itaconate, which have been shown to impair NLRP3 inflammasome activation and IL-1β release in blood mononuclear cells from CAPS patients. In this study, we provide insight into the inhibitory mechanisms by which the itaconate derivative 4-octyl itaconate (4-OI) acts on NLRP3 that has different gain-of-function mutations (p.R262W, p.D305N and p.T350M) associated with CAPS. Notably, 4-OI effectively blocks the basal auto-activation of the inflammasome formed by NLRP3 p.R262W, p.D305N and p.T350M variants, which in turn reduces caspase-1 activation, gasdermin D processing, and IL-18 release. Furthermore, after lipopolysaccharide priming of macrophages, 4-OI also decreases IL-1β gene expression and release. Overall, 4-OI impairs CAPS-associated inflammasome function at multiple levels, meaning that therapeutic agents based on itaconate could be a promising therapeutic approach to managing inflammatory episodes in CAPS patients carrying p.R262W, p.D305N or p.T350M variants.
Collapse
Affiliation(s)
- Cristina Molina-Lopez
- Molecular Inflammation Group, Biomedical Research Institute of Murcia (IMIB), Edificio LAIB 4ª Planta, Carretera Buenavista S/N, 30120, El Palmar, Murcia, Spain
- CABIMER, Seville, Spain
| | - Laura Hurtado-Navarro
- Molecular Inflammation Group, Biomedical Research Institute of Murcia (IMIB), Edificio LAIB 4ª Planta, Carretera Buenavista S/N, 30120, El Palmar, Murcia, Spain
- IdiPaz, Madrid, Spain
| | - Luke A J O'Neill
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Pablo Pelegrin
- Molecular Inflammation Group, Biomedical Research Institute of Murcia (IMIB), Edificio LAIB 4ª Planta, Carretera Buenavista S/N, 30120, El Palmar, Murcia, Spain.
- Department of Biochemistry and Molecular Biology B and Immunology, Faculty of Medicine, University of Murcia, 30120, Murcia, Spain.
| |
Collapse
|
4
|
Ogunware AE, Kielian T. Immunometabolism shapes chronic Staphylococcus aureus infection: insights from biofilm infection models. Curr Opin Microbiol 2025; 86:102612. [PMID: 40409167 DOI: 10.1016/j.mib.2025.102612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 04/17/2025] [Accepted: 04/28/2025] [Indexed: 05/25/2025]
Abstract
Staphylococcus aureus is both a commensal bacterium and versatile pathogen, capable of transitioning from a benign colonizer to cause invasive disease. Its ability to form biofilm - a resilient, highly structured bacterial community - plays a key role in chronic infections, including those associated with medical implants and native tissues. The unique microenvironments of these biofilm niches create challenges for the host immune system, complicating pathogen clearance. Immunometabolism, the interplay between immune function and metabolic programming, plays a crucial role in dictating how the host combats S. aureus biofilms. Leukocytes undergo profound metabolic changes in response to biofilm, which can lead to dysregulated immune responses and persistent infection. This review explores recent insights defining the metabolic landscape of immune responses to S. aureus biofilm with a focus on two clinically relevant models, namely, craniotomy and prosthetic joint infection.
Collapse
Affiliation(s)
- Adedayo E Ogunware
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Tammy Kielian
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| |
Collapse
|
5
|
Yang X, Deng Y, Ye Y, Meng J, Su M, Wei W, Qin Y, Zhang H, Tian Y, Deng S, Liao Z, Zhou Z, Li J, Hu Y, Zhang B, Sun Y, Wen L, Zhang Z, Huang F, Wan C, Yang K. Macrophage-Derived Itaconate Suppresses Dendritic Cell Function to Promote Acquired Resistance to Anti-PD-1 Immunotherapy. Cancer Res 2025; 85:1842-1856. [PMID: 40036156 DOI: 10.1158/0008-5472.can-24-2982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 01/07/2025] [Accepted: 02/26/2025] [Indexed: 03/06/2025]
Abstract
Adaptive resistance to immunotherapy remains a significant challenge in cancer treatment. The reshaping of the tumor immune microenvironment in response to therapeutic pressures is a crucial factor contributing to this resistance. In this study, by comprehensive metabolic profiling of tumor tissues, we identified elevated itaconate in response to anti-PD-1 therapy as an adaptive resistance mechanism that promoted immune escape and tumor progression. CD8+ T-cell-derived IFNγ induced a significant upregulation of cis-aconitate decarboxylase 1 (ACOD1) in macrophages via the JAK-STAT1 pathway, thereby rewiring the Krebs cycle toward itaconate production. In murine models, macrophage-specific deletion of Acod1 increased the antitumor efficacy of anti-PD-1 therapy and improved survival. Additionally, itaconate and its derivative, 4-octyl itaconate, suppressed the tumor antigen presentation and cross-priming ability of dendritic cells, resulting in the impairment of antigen-specific T-cell antitumor responses. In summary, these findings identify an IFNγ-dependent immunometabolic mechanism of anti-PD-1 resistance, providing a promising strategy for combination therapy. Significance: Elevated itaconate production by macrophages induced by IFNγ is a critical negative feedback immunoregulatory metabolic response to anti-PD-1 immunotherapy that inhibits the cross-priming function of dendritic cells and confers immunotherapy resistance.
Collapse
Affiliation(s)
- Xiao Yang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China
| | - Yue Deng
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China
| | - Ying Ye
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China
| | - Jingshu Meng
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China
| | - Mengyao Su
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China
| | - Wenwen Wei
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China
| | - You Qin
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China
| | - Haibo Zhang
- Cancer Center, Department of Radiation Oncology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, China
| | - Yu Tian
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China
| | - Suke Deng
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China
| | - Zhiyun Liao
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China
| | - Zhiyuan Zhou
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China
| | - Jie Li
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China
| | - Yan Hu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China
| | - Bin Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China
| | - Yajie Sun
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China
| | - Lu Wen
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China
| | - Zhanjie Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China
| | - Fang Huang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China
| | - Chao Wan
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China
| | - Kunyu Yang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China
| |
Collapse
|
6
|
Xu P, Chen X, Li Q, Dong Z, Zhu J, Su Z, Zhang Q, Fang K. Nontargeted metabolomics uncovering metabolite signatures in glioblastoma: a preliminary study on candidate biomarker discovery for IDH subtyping and survival prediction. Front Oncol 2025; 15:1568040. [PMID: 40406272 PMCID: PMC12095168 DOI: 10.3389/fonc.2025.1568040] [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: 01/28/2025] [Accepted: 04/16/2025] [Indexed: 05/26/2025] Open
Abstract
Background Currently, there are no established tumor-derived metabolic biomarkers in clinical practice that can simultaneously differentiate among nontumorous brain tissues, isocitrate dehydrogenase (IDH) wild-type glioblastomas (GBMs), and IDH mutant GBMs, or accurately predict patient survival. The aim of this study was to identify GBM biomarkers for molecular classification and survival prediction via nontargeted metabolomics. Methods Brain tissue samples from nontumors, IDH-mutant GBMs, and IDH-wild-type GBMs were analyzed via liquid chromatography-mass spectrometry (LC-MS). Metabolites for molecular classification and survival prediction were identified via sparse partial least-squares discriminant analysis (sPLS-DA) and extreme gradient boosting (XGBoost) models, respectively. Both sets of metabolites were then validated via bootstrap resampling. The biomarkers for survival prediction were further validated using an independent metabolomics dataset. Results In total, 185 human-derived metabolites were identified with high confidence levels. Two non-overlapping sets of 11 candidate biomarkers for molecular subtyping and survival prediction were screened out. In the validation models for molecular subtyping, the random forest model achieved the highest accuracy (0.787, 95% CI: 0.780-0.795) and a Kappa value of 0.681. The Cox proportional hazards regression model established based on cholic acid and citrulline had an AUC of 0.942 (95% CI: 0.920-0.956) at 84 days and an AUC of 0.812 (95% CI: 0.746-0.826) at 297 days. Conclusion This exploratory study identified potential metabolic biomarkers for GBM subtyping and prognosis prediction. However, further validation in large-scale clinical studies and mechanistic investigations are needed to confirm their applicability and reliability.
Collapse
Affiliation(s)
- Peng Xu
- Clinical Laboratory, The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Xiling Chen
- Clinical Laboratory, The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Qun Li
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zheqing Dong
- Clinical Laboratory, The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Ji Zhu
- Clinical Laboratory, The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhipeng Su
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Qifan Zhang
- Clinical Laboratory, The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Kui Fang
- Clinical Laboratory, The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
7
|
Yuan YS, Li HY, Lu H, Li GC, Cao Z, Xu C, Xiao HH, Zhang LP, Xu HL. Reprogramming mitochondrial metabolism to enhance macrophages polarization by ROS-responsive nanoparticles for osteoarthritis. Biomaterials 2025; 322:123395. [PMID: 40403559 DOI: 10.1016/j.biomaterials.2025.123395] [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: 12/31/2024] [Revised: 04/28/2025] [Accepted: 05/06/2025] [Indexed: 05/24/2025]
Abstract
Osteoarthritis (OA) is a chronic low-grade inflammatory joint disease closely related to the inflammatory pathological microenvironment caused by synovial M1 macrophages. In contrast to the proinflammatory role of M1 macrophages, M2 macrophages contribute to anti-inflammatory responses and tissue repair. Therefore, shifting the M2/M1 phenotype ratio in favor of M2 macrophages has become a promising therapeutic strategy for OA. However, current therapeutics cannot penetrate the synovium and only show limited drug retention time. Herein, we developed an OA microenvironment-responsive nanocarrier with thioketal bonds in the main chain and β-1,3-d-glucan and triphenylphosphine units in the side chain, which can respond to reactive oxygen species (ROS) and target macrophages and mitochondrial aggregation. For OA treatment, 4-octyl itaconate and dexamethasone were encapsulated within the nanocarrier, forming HBPTG@OD that effectively eliminated mitochondrial ROS and inducible nitric oxide synthase in M1 macrophages. HBPTG@OD significantly suppressed the release of inflammatory factors by macrophages and thereby reducing chondrocyte death. In vivo studies in a destabilized medial meniscus (DMM)-induced OA model showed that HBPTG@OD effectively converted M1 synovial macrophages to M2 macrophages, consequently delaying chondrogenic apoptosis. This study presents a nanocarrier-based strategy that effectively repolarizes M1 macrophages, demonstrating great promise for the treatment of OA.
Collapse
Affiliation(s)
- Yu-Song Yuan
- Department of Trauma and Orthopedics, Foot and Ankle Center, Peking University People's Hospital, 11th Xizhimen South Street, Beijing, China, 100044; Department of Trauma and Orthopedics, China-Japan Friendship Hospital, 2nd Yinghuayuan East Street, Beijing, China, 100029
| | - Hui-Yun Li
- Department of Orthopedic Surgery, The First Affiliated Hospital of University of South China, Hengyang, 421001, Hunan, China; Hunan Key Laboratory of Joint Degeneration and Injury, Xiangya Hospital, Central South University, Changsha, China
| | - Hao Lu
- Department of Trauma and Orthopedics, Foot and Ankle Center, Peking University People's Hospital, 11th Xizhimen South Street, Beijing, China, 100044
| | - Gui-Cheng Li
- Department of Trauma and Orthopedics, Foot and Ankle Center, Peking University People's Hospital, 11th Xizhimen South Street, Beijing, China, 100044
| | - Zheng Cao
- Department of Chemical and Biomolecular Engineering, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Chun Xu
- School of Dentistry, The University of Queensland, Brisbane, 4006, Australia
| | - Hai-Hua Xiao
- Beijing National Laboratory for Molecular Science, State Key Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Science, Beijing, 100190, China.
| | - Ling-Pu Zhang
- Beijing National Laboratory for Molecular Science, State Key Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Science, Beijing, 100190, China.
| | - Hai-Lin Xu
- Department of Trauma and Orthopedics, Foot and Ankle Center, Peking University People's Hospital, 11th Xizhimen South Street, Beijing, China, 100044.
| |
Collapse
|
8
|
Hao D, McBride MA, Bohannon JK, Hernandez A, Klein B, Williams DL, Sherwood ER. Metabolic adaptations driving innate immune memory: mechanisms and therapeutic implications. J Leukoc Biol 2025; 117:qiaf037. [PMID: 40138361 DOI: 10.1093/jleuko/qiaf037] [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: 12/20/2024] [Revised: 03/12/2025] [Accepted: 03/24/2025] [Indexed: 03/29/2025] Open
Abstract
Immune memory is a hallmark of the adaptive immune system. However, recent research reveals that innate immune cells also retain memory of prior pathogen exposure that prompts enhanced responses to subsequent infections. This phenomenon is termed "innate immune memory" or "trained immunity." Notably, remodeling of cellular metabolism, which closely links to epigenetic reprograming, is a prominent feature of innate immune memory. Adaptations in glycolysis, the tricarboxylic acid cycle, oxidative phosphorylation, glutaminolysis, and lipid synthesis pathways are critical for establishing innate immune memory. This review provides an overview of the current understanding of how metabolic adaptations drive innate immune memory. This understanding is fundamental to understanding innate immune system functions and advancing therapies against infectious diseases.
Collapse
Affiliation(s)
- Dan Hao
- Department of Anesthesiology, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville, TN 37232, United States
| | - Margaret A McBride
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville, TN 37232, United States
| | - Julia K Bohannon
- Department of Anesthesiology, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville, TN 37232, United States
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville, TN 37232, United States
| | - Antonio Hernandez
- Department of Anesthesiology, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville, TN 37232, United States
| | - Benjamin Klein
- Department of Anesthesiology, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville, TN 37232, United States
| | - David L Williams
- Department of Surgery, East Tennessee State University, Quillen College of Medicine, P.O. Box 70575, Johnson City, TN 37614, United States
- Center for Inflammation, Infectious Disease and Immunology, Quillen College of Medicine, 1276 Gilbreath Drive, Johnson City, TN 37614, United States
| | - Edward R Sherwood
- Department of Anesthesiology, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville, TN 37232, United States
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville, TN 37232, United States
- Department of Surgery, East Tennessee State University, Quillen College of Medicine, P.O. Box 70575, Johnson City, TN 37614, United States
- Center for Inflammation, Infectious Disease and Immunology, Quillen College of Medicine, 1276 Gilbreath Drive, Johnson City, TN 37614, United States
| |
Collapse
|
9
|
Pålsson-McDermott EM, O'Neill LAJ. Gang of 3: How the Krebs cycle-linked metabolites itaconate, succinate, and fumarate regulate macrophages and inflammation. Cell Metab 2025; 37:1049-1059. [PMID: 40169002 DOI: 10.1016/j.cmet.2025.03.004] [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: 01/30/2025] [Revised: 02/18/2025] [Accepted: 03/05/2025] [Indexed: 04/03/2025]
Abstract
The reprogramming of metabolic pathways and processes in immune cells has emerged as an important aspect of the immune response. Metabolic intermediates accumulate as a result of metabolic adaptations and mediate functions outside of metabolism in the regulation of immunity and inflammation. In macrophages, there has been a major focus on 3 metabolites linked to the Krebs cycle, itaconate, succinate, and fumarate, which have been shown to regulate multiple processes. Here, we discuss recent progress on these 3 metabolites with regard to their effect on macrophages in host defense and inflammatory diseases. We also consider the therapeutic opportunities presented from the mimicry of these metabolites or by targeting the enzymes that make or metabolize them in order to leverage the body's own anti-inflammatory response.
Collapse
Affiliation(s)
- Eva M Pålsson-McDermott
- School of Biochemistry and Immunology, Trinity Biomedical Science Institute, Trinity College Dublin, Dublin 2, Ireland.
| | - Luke A J O'Neill
- School of Biochemistry and Immunology, Trinity Biomedical Science Institute, Trinity College Dublin, Dublin 2, Ireland
| |
Collapse
|
10
|
El-Derany MO, Ramakrishnan SK, Li Y, Buscher K, Jarad CA, Schaller ML, Cantwell M, Vigil TM, Frieler RA, Sajjakulnukit P, Lyssiotis CA, Mortensen RM, Shah YM. Itaconate potentiates hepatic gluconeogenesis through NRF2 induction. PLoS One 2025; 20:e0322946. [PMID: 40323920 PMCID: PMC12052187 DOI: 10.1371/journal.pone.0322946] [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: 12/24/2024] [Accepted: 03/31/2025] [Indexed: 05/07/2025] Open
Abstract
The interplay between systemic metabolism and immune responses is increasingly recognized as a significant factor in the dysregulation of glucose homeostasis associated with diabetes and obesity. Immune metabolites play crucial roles in mediating this crosstalk, with itaconate emerging as an important immune metabolite involved in the inflammatory response of macrophages. Recent studies have highlighted the role of itaconate as a regulator of glucose metabolism, particularly in the context of obesity, although the underlying mechanisms remain poorly understood. In this study, we identified itaconate as one of the metabolites that significantly increase in the liver during fasting compared to fed conditions. Mechanistically, we found that itaconate enhances glucagon-induced liver gluconeogenesis independently of insulin signaling. Notably, itaconate upregulates the expression of gluconeogenic genes both under basal conditions and in the presence of palmitic acid. Furthermore, our data indicate that the effects of itaconate occur independently of CREB activation. Instead, we demonstrate that these potentiating effects are mediated through the induction of nuclear factor erythroid 2-related factor 2 (NRF2). Our findings demonstrate that itaconate has a glucagon-potentiating effects in the liver, suggesting that itaconate may play a significant role in the pathogenesis of metabolic-associated liver diseases.
Collapse
Affiliation(s)
- Marwa O. El-Derany
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, United States of America
- Department of Biochemistry, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Sadeesh K. Ramakrishnan
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Yingjie Li
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Kathryn Buscher
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Christina A. Jarad
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Megan L. Schaller
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Marc Cantwell
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Thomas M. Vigil
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Ryan A. Frieler
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Peter Sajjakulnukit
- University of Michigan Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Costas A. Lyssiotis
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, United States of America
- University of Michigan Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan, United States of America
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Richard M. Mortensen
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Yatrik M. Shah
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, United States of America
- University of Michigan Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan, United States of America
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
| |
Collapse
|
11
|
Woodworth KE, Froom ZSCS, Osborne ND, Rempe CN, Wheeler B, Medd K, Callaghan NI, Qian H, Acharya AP, Charron C, Davenport Huyer L. Development of Itaconate Polymers Microparticles for Intracellular Regulation of Pro-Inflammatory Macrophage Activation. Adv Healthc Mater 2025; 14:e2405257. [PMID: 40183748 DOI: 10.1002/adhm.202405257] [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: 12/27/2024] [Revised: 03/12/2025] [Indexed: 04/05/2025]
Abstract
Itaconate (IA) is an endogenous metabolite and a potent regulator of the innate immune system. It's use in immunomodulatory therapies has faced limitations due to challenges in controlled delivery and requirements of high extracellular concentrations for internalization of the highly polar small molecule to achieve its intracellular therapeutic activity. Microparticle (MP)-based delivery strategies are a promising approach for intracellular delivery of small molecule metabolites through macrophage phagocytosis and subsequent intracellular polymer degradation-based delivery. Toward the goal of intracellular delivery of IA, degradable polyester polymer- (poly(dodecyl itaconate)) based IA polymer microparticles (IA-MPs) are generated using an emulsion method, forming micron-scale (≈1.5 µm) degradable microspheres. IA-MPs are characterized with respect to their material properties and IA release kinetics to inform particle fabrication. Treatment of murine bone marrow-derived macrophages with an optimized particle concentration of 0.1 mg million-1 cells enables phagocytosis-mediated internalization and low levels of cytotoxicity. Flow cytometry demonstrates IA-MP-specific regulation of IA-sensitive inflammatory targets. Metabolic analyses demonstrate that IA-MP internalization inhibits oxidative metabolism and induced glycolytic reliance, consistent with the established mechanism of IA-associated inhibition of succinate dehydrogenase. This development of IA-based polymer microparticles provides a basis for additional innovative metabolite-based microparticle drug delivery systems for the treatment of inflammatory disease.
Collapse
Affiliation(s)
- Kaitlyn E Woodworth
- School of Biomedical Engineering, Faculties of Medicine and Engineering, Dalhousie University, Halifax, NS, B3H 4R2, Canada
| | - Zachary S C S Froom
- School of Biomedical Engineering, Faculties of Medicine and Engineering, Dalhousie University, Halifax, NS, B3H 4R2, Canada
| | - Natasha D Osborne
- Department of Microbiology & Immunology, Faculty of Medicine, Dalhousie University, Halifax, NS, B3H 4R2, Canada
| | - Christian N Rempe
- Department of Microbiology & Immunology, Faculty of Medicine, Dalhousie University, Halifax, NS, B3H 4R2, Canada
| | - Brenden Wheeler
- School of Biomedical Engineering, Faculties of Medicine and Engineering, Dalhousie University, Halifax, NS, B3H 4R2, Canada
| | - Kyle Medd
- School of Biomedical Engineering, Faculties of Medicine and Engineering, Dalhousie University, Halifax, NS, B3H 4R2, Canada
| | - Neal I Callaghan
- Faculty of Medicine, Dalhousie University, Halifax, NS, B3H 4R2, Canada
| | - Huikang Qian
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Abhinav P Acharya
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Carlie Charron
- Department of Chemistry, Faculty of Science, Dalhousie University, Halifax, NS, B3H 4R2, Canada
| | - Locke Davenport Huyer
- School of Biomedical Engineering, Faculties of Medicine and Engineering, Dalhousie University, Halifax, NS, B3H 4R2, Canada
- Department of Microbiology & Immunology, Faculty of Medicine, Dalhousie University, Halifax, NS, B3H 4R2, Canada
- Department of Biomaterials & Applied Oral Sciences, Faculty of Dentistry, Dalhousie University, Halifax, NS, B3H 4R2, Canada
- Department of Surgery, Nova Scotia Health, Halifax, NS, B3H 4R2, Canada
| |
Collapse
|
12
|
Prenen F, Pollenus E, Meers H, Knoops S, Sadler R, Deckers M, Mills EL, Van den Steen PE. Itaconate Has Limited Protective Effects in Experimental Malaria Models. Eur J Immunol 2025; 55:e202451595. [PMID: 40346757 DOI: 10.1002/eji.202451595] [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: 10/22/2024] [Revised: 04/24/2025] [Accepted: 04/25/2025] [Indexed: 05/12/2025]
Abstract
In severe malaria, dysregulated metabolism and excessive inflammatory responses contribute to fatal outcomes. Therapeutic strategies that address both metabolic and inflammatory balances are thus required. Itaconate, a metabolite produced by aconitate decarboxylase 1 (ACOD1), is a potent inhibitor of both inflammation and glycolysis with protective effects in various inflammatory diseases. Although elevated itaconate levels have been observed in Plasmodium-infected individuals, its role in malaria is still poorly understood, making further investigation essential for assessing its therapeutic potential. We investigated the role of itaconate in both severe and mild malaria using Plasmodium berghei NK65 (PbNK65) and Plasmodium chabaudi AS (PcAS) models, respectively. Using 13C-tracer metabolomics, we detected increased itaconate levels in various organs during infection and identified inflammatory monocytes as the source of this production. Nevertheless, ACOD1 knockout mice displayed no significant changes in phenotype after PbNK65 infection, and treatment of PbNK65-infected mice with 4-octyl itaconate did not affect disease severity either. However, in the PcAS model, ACOD1 deficiency worsened the disease, as indicated by increased weight loss, higher clinical scores, and elevated parasitemia. Therefore, in contrast to the findings in recent literature, our study shows that itaconate does not contribute to susceptibility, but rather provides limited protection to malaria.
Collapse
Affiliation(s)
- Fran Prenen
- Laboratory of Immunoparasitology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Emilie Pollenus
- Laboratory of Immunoparasitology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Hanne Meers
- Laboratory of Immunoparasitology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
- Laboratory of Immunobiology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Sofie Knoops
- Laboratory of Immunoparasitology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Rebecca Sadler
- Laboratory of Immunoparasitology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Margot Deckers
- Laboratory of Immunoparasitology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
- Laboratory of Host-Pathogen Interactions, Department of Pathology, University of Utah, Salt Lake City, USA
| | - Evanna L Mills
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Immunology, Harvard Medical School, Boston, Massachusetts, USA
| | - Philippe E Van den Steen
- Laboratory of Immunoparasitology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| |
Collapse
|
13
|
Wang T, Pan R, Wen J, Ma X. Dexmedetomidine modulates peritoneal macrophage to attenuate lipopolysaccharide-induced inflammation. Cell Immunol 2025; 411-412:104942. [PMID: 40088848 DOI: 10.1016/j.cellimm.2025.104942] [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: 11/22/2024] [Revised: 03/04/2025] [Accepted: 03/10/2025] [Indexed: 03/17/2025]
Abstract
PURPOSE To investigate how Dexmedetomidine (Dex) modulates the function of peritoneal macrophages (PMs) to reduce lipopolysaccharide (LPS)-induced inflammation. METHODS The anti-inflammatory effect of Dex on LPS-stimulated PMs was assessed by examining its impact on their proliferation, phagocytosis, and polarization. Proliferation and phagocytic activity were measured using CCK-8 and Neutral Red staining assays, respectively. The levels of inflammatory mediators were quantified using ELISA. Additionally, macrophage polarization was evaluated via ELISA, flow cytometry, and Western blot analysis to identify shifts in macrophage phenotypes. RESULTS Dex increased the proliferation and phagocytic capabilities of PMs, thereby mitigating LPS-induced inflammation. It suppressed pro-inflammatory mediators, including tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), and high mobility group box 1 (HMGB1), while increasing levels of the anti-inflammatory cytokine interleukin-10 (IL-10). Furthermore, Dex promoted M2-type macrophage polarization, characterized by increased expression of IL-10, CD206, Arg-1, and CD11c. This effect was mediated through the JAK1/STAT6 signaling pathway, promoting M2 polarization, which was attenuated when JAK1 and STAT6 expression were downregulated. CONCLUSION Dex reduces LPS-induced inflammation in part by enhancing the proliferation, phagocytosis, and M2 polarization of PMs, with a key role for the JAK1/STAT6 pathway in promoting anti-inflammatory responses during sepsis.
Collapse
Affiliation(s)
- Tao Wang
- Guiqian International General Hospital, Department of Critical Care Medicine, Guiyang, City, Guizhou Province, China; Department of Critical Care Medicine, Zuiyi First People's Hospital (The Third Affiliated Hospital of Zuiyi Medical University), Zuiyi City,Guizhou Province, China
| | - Rui Pan
- Department of Critical Care Medicine, Zuiyi First People's Hospital (The Third Affiliated Hospital of Zuiyi Medical University), Zuiyi City,Guizhou Province, China
| | - Jianli Wen
- Department of Critical Care Medicine, Zuiyi First People's Hospital (The Third Affiliated Hospital of Zuiyi Medical University), Zuiyi City,Guizhou Province, China
| | - Xinglong Ma
- Department of Critical Care Medicine, Zuiyi First People's Hospital (The Third Affiliated Hospital of Zuiyi Medical University), Zuiyi City,Guizhou Province, China.
| |
Collapse
|
14
|
Zhu B, Zhu L, Ge Z, Zheng S, Dai X, Feng D, Tan L, Sha P, Yao Y. 4-Octyl Itaconate Modulates Dendritic Cells Function and Tumor Immunity via NRF2 Activation. J Inflamm Res 2025; 18:5699-5713. [PMID: 40322530 PMCID: PMC12047388 DOI: 10.2147/jir.s516085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 04/22/2025] [Indexed: 05/08/2025] Open
Abstract
Objective Dendritic cells (DCs) play a pivotal role in orchestrating anti-tumor immune responses. However, various factors can suppress DCs function and compromise anti-tumor immunity. Itaconate, a metabolite activated during inflammation and infection, has been identified to possess immunomodulatory properties, but its role on DCs remains largely unexplored. In this study, we aimed to investigate the role of itaconate in regulating the maturation and function of DCs and its underlying molecular mechanism. Methods Bone marrow-derived dendritic cells (BMDCs) were treated with 4-octyl itaconate (4OI). The expression levels of CD40, CD80, CD86, and MHC-II on BMDCs were analyzed by flow cytometry. The mRNA expression of cytokines was assessed using RT-qPCR. BMDCs with different treatment were adoptively transferred to B16-OVA tumor-bearing mice. The production of IFN-γ, IL-2, and TNF-α in CD4+ T and CD8+ T cells were analyzed by flow cytometry. The protein level of NRF2 in BMDCs was analyzed by Western blot. Results Treatment with 4OI represses DC maturation and function. Specifically, 4OI-treated DCs exhibited impaired phenotypic and functional maturation, characterized by decreased expression of co-stimulatory molecules CD40, CD80, and CD86, as well as lower levels of pro-inflammatory cytokines IL-12, IL-6, TNF-α and IL-1β. Furthermore, these DCs demonstrated a diminished capacity to stimulate T cell responses both in vitro and in vivo. Mechanistically, 4OI inhibits DCs maturation and function through enhancing and activating KEAP1/NRF2 pathway. Conclusion This study reveals that 4OI inhibits DC function through NRF2 activation, elucidating the immunomodulatory mechanisms of itaconate and emphasizing its pivotal role in developing targeted DC-based tumor immunotherapy strategies.
Collapse
Affiliation(s)
- Bo Zhu
- Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University; Institute of Neuroscience and Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, Jiangsu Province, 215000, People’s Republic of China
- Department of Laboratory Medicine, Institute of Medical Immunology of Jiangsu University, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu Province, 215123, People’s Republic of China
- Institutes of Biology and Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, Jiangsu Province, 215000, People’s Republic of China
| | - Lihua Zhu
- Department of Laboratory Medicine, Institute of Medical Immunology of Jiangsu University, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu Province, 215123, People’s Republic of China
| | - Zongxia Ge
- Department of Laboratory Medicine, Institute of Medical Immunology of Jiangsu University, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu Province, 215123, People’s Republic of China
| | - Songhang Zheng
- Department of Laboratory Medicine, Institute of Medical Immunology of Jiangsu University, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu Province, 215123, People’s Republic of China
| | - Xiaoqiu Dai
- Institutes of Biology and Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, Jiangsu Province, 215000, People’s Republic of China
| | - Dingqi Feng
- Department of Central Laboratory, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu Province, 215123, People’s Republic of China
- Center of Clinical Laboratory, Dushu Lake Hospital Affiliated to Soochow University, Suzhou, Jiangsu Province, 215000, People’s Republic of China
| | - Lu Tan
- Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University; Institute of Neuroscience and Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, Jiangsu Province, 215000, People’s Republic of China
| | - Pengfei Sha
- Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University; Institute of Neuroscience and Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, Jiangsu Province, 215000, People’s Republic of China
| | - Yizheng Yao
- Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University; Institute of Neuroscience and Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, Jiangsu Province, 215000, People’s Republic of China
| |
Collapse
|
15
|
Chen X, Zhu Y, Xia L, Su S, Fan S, Lu Y, Chen Q, Wei Y, Huang Q, Liu X, Peng X. Glutamine limits NLRP3 inflammasome activation and pyroptosis in macrophages by sustaining the IRG1/itaconate axis. FEBS J 2025. [PMID: 40296302 DOI: 10.1111/febs.70119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 01/18/2025] [Accepted: 04/17/2025] [Indexed: 04/30/2025]
Abstract
Aberrant activation of NACHT, LRR, and PYD domains-containing protein 3 (NLRP3) inflammasome increases the release of mature pro-inflammatory cytokines interleukin (IL)-1β and IL-18, and enhances pyroptosis; thereby necessitating tight regulation of the NLRP3 inflammasome. Dysfunctional glutamine metabolism contributes to the pathogenesis of multiple inflammatory disorders, and the precise mechanism remains to be elucidated. Here, we provide evidence that glutamine deprivation enhances NLRP3 inflammasome activation in macrophages. Indeed, the absence of exogenous glutamine specifically enhanced NLRP3 inflammasome assembly, thereby accelerating pyroptosis and promoting the maturation of IL-1β and IL-18. Inhibition of glutaminolysis exhibited a similar effect to glutamine deprivation, whereas this effect was reversed by α-ketoglutarate (α-KG), a tricarboxylic acid (TCA)-cycle intermediate that can be replenished by glutamine supply. We further observed reduced generation of endogenous itaconate by glutamine deprivation and verified that both exogenous supplementation of itaconate derivative and increased endogenous itaconate production by overexpressing immune-responsive gene 1 [IRG1; also known as aconitate decarboxylase 1 (ACOD1)] could replace glutamine to inhibit the NLRP3 inflammasome. Mechanistically, glutamine deprivation decreased the source of substrate and inhibited transcription factor EB (TFEB)-dependent transcriptional upregulation of IRG1, thereby impairing the IRG1/itaconate axis that suppresses the NLRP3 inflammasome. Furthermore, glutamine deficiency was detected in a murine sepsis model, whereas extrinsic glutamine supplementation conferred protection against intestinal inflammation and tissue damage in septic mice. Taken together, our findings provide a novel insight into the link between glutamine metabolism and NLRP3 inflammasome activation, highlighting the target of glutamine metabolism, which holds as a potential therapeutic strategy for inflammatory diseases.
Collapse
Affiliation(s)
- Xiaoli Chen
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yuanfeng Zhu
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Lin Xia
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Sen Su
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Shijun Fan
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yongling Lu
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Qian Chen
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yan Wei
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Qianying Huang
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Xin Liu
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Xi Peng
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- State Key Laboratory of Trauma and Chemical Poisoning, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| |
Collapse
|
16
|
Ramalho T, Gazzinelli RT. Itaconate boosts type I IFN response by disrupting cytoprotection. Nat Metab 2025:10.1038/s42255-025-01274-1. [PMID: 40251411 DOI: 10.1038/s42255-025-01274-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/20/2025]
Affiliation(s)
- Theresa Ramalho
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA.
| | - Ricardo Tostes Gazzinelli
- Department of Medicine, Division of Infectious Disease and Immunology, University of Massachusetts Chan Medical School, Worcester, MA, USA.
- Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.
- Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Brazil.
| |
Collapse
|
17
|
Qiu C, Zhang W, Zhao Y, Han T, Yang W, Liu Y, Jin P, Chen J, Shuai X, Ren J, Huang P. Reprogramming Glucose Metabolism of Macrophage for Acute Liver Failure Therapy with Itaconate Lipo-Nanodrug. Adv Healthc Mater 2025:e2500019. [PMID: 40249158 DOI: 10.1002/adhm.202500019] [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: 01/02/2025] [Revised: 03/17/2025] [Indexed: 04/19/2025]
Abstract
Acute liver failure (ALF) is a life-threatening disease featuring comprehensive inflammatory response and metabolic disorders in which macrophages exert central roles. A glucose metabolism mediator of macrophages, itaconate, has demonstrated potent anti-inflammatory efficacy in various diseases, implying that itaconate could work in treating ALF. However, systemic administration of itaconate may lead to immune disorder, making targeting the delivery of itaconate to the liver lesion area highly important. Herein, a liposomal nanodrug incorporating itaconate is developed, and its potential in treating acute liver failure in an ALF murine model established by LPS/D-GalN administration is tested. The nanodrug shows preferential liver accumulation to effectively alleviate LPS/D-GalN-induced hepatic histopathological injury by decreasing oxidative stress. Moreover, it reprograms the glucose metabolism of macrophages, resulting in macrophage repolarization toward the anti-inflammatory phenotype. Furthermore, western-blot and immunohistochemical assays verifies that the nanodrug may inhibit aerobic glycolysis of macrophages in an NRF2 and STING-dependent manner. These results underline the promise of the nanodrug for ALF treatment by reprogramming glucose metabolism.
Collapse
Affiliation(s)
- Chen Qiu
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Wei Zhang
- Department of Medical Ultrasound, Laboratory of Novel Optoacoustic (Ultrasonic) Imaging, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
| | - Yanan Zhao
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Tian Han
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Wende Yang
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Yajing Liu
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Peile Jin
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Jifan Chen
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Xintao Shuai
- Nanomedicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Jie Ren
- Department of Medical Ultrasound, Laboratory of Novel Optoacoustic (Ultrasonic) Imaging, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
| | - Pintong Huang
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Research Center for Life Science and Human Health, Binjiang Institute of Zhejiang University, Hangzhou, 310009, China
| |
Collapse
|
18
|
Wang S, Qin L, Liu F, Zhang Z. Unveiling the crossroads of STING signaling pathway and metabolic reprogramming: the multifaceted role of the STING in the TME and new prospects in cancer therapies. Cell Commun Signal 2025; 23:171. [PMID: 40197235 PMCID: PMC11977922 DOI: 10.1186/s12964-025-02169-0] [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: 01/26/2025] [Accepted: 03/23/2025] [Indexed: 04/10/2025] Open
Abstract
The cGAS-STING signaling pathway serves as a critical link between DNA sensing and innate immunity, and has tremendous potential to improve anti-tumor immunity by generating type I interferons. However, STING agonists have shown decreasing biotherapeutic efficacy in clinical trials. Tumor metabolism, characterized by aberrant nutrient utilization and energy production, is a fundamental hallmark of tumorigenesis. And modulating metabolic pathways in tumor cells has been discovered as a therapeutic strategy for tumors. As research concerning STING progressed, emerging evidence highlights its role in metabolic reprogramming, independent its immune function, indicating metabolic targets as a strategy for STING activation in cancers. In this review, we delve into the interplay between STING and multiple metabolic pathways. We also synthesize current knowledge on the antitumor functions of STING, and the metabolic targets within the tumor microenvironment (TME) that could be exploited for STING activation. This review highlights the necessity for future research to dissect the complex metabolic interactions with STING in various cancer types, emphasizing the potential for personalized therapeutic strategies based on metabolic profiling.
Collapse
Affiliation(s)
- Siwei Wang
- Hepatic Surgery Center, Hubei Province for the Clinical Medicine Research Center of Hepatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, 430030, Hubei, China
- Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Lu Qin
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation, Ministry of Education, Huazhong University of Science and Technology), Wuhan, China
| | - Furong Liu
- Hepatic Surgery Center, Hubei Province for the Clinical Medicine Research Center of Hepatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China.
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, 430030, Hubei, China.
- Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China.
| | - Zhanguo Zhang
- Hepatic Surgery Center, Hubei Province for the Clinical Medicine Research Center of Hepatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China.
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, 430030, Hubei, China.
- Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China.
| |
Collapse
|
19
|
Ma J, Ao Y, Yue Z, Wang Z, Hou X, Li H, Wang H, Luo S, He J, Duan Z, Liu L, Wei K. Elevated GFI1 in Alveolar Macrophages Suppresses ACOD1 Expression and Exacerbates Lipopolysaccharide-Induced Lung Injury in Obesity. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2413546. [PMID: 39921443 PMCID: PMC11967830 DOI: 10.1002/advs.202413546] [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: 01/13/2025] [Indexed: 02/10/2025]
Abstract
To investigate the mechanisms behind the worsening of acute lung injury (ALI) in obesity, transcriptomic sequencing is performed, and significantly reduced mRNA levels of Aconitate Decarboxylase 1 (ACOD1) in the lung tissue of high-fat diet (HFD) mice are found. Clinical samples are collected, an ALI model is established in HFD mice, and both human and mouse samples are analyzed, revealing a significant decrease in ACOD1 expression in lung tissue and alveolar macrophages in obesity. Further in vivo and in vitro experiments show that ACOD1 knockdown worsens lung injury, inflammation, and oxidative stress, while ACOD1 overexpression alleviates these effects. Moreover, nuclear factor erythroid 2-related factor 2 (Nrf2) inhibition diminishes the protective effects of ACOD1 overexpression in ALI exacerbated by obesity. Additionally, in the context of obesity, growth factor independent 1 (GFI1) protein levels are elevated in alveolar macrophages, and its knockdown leads to upregulated ACOD1 expression. Therefore, this study suggests that ACOD1 downregulation in alveolar macrophages is a key factor in worsening ALI in obesity, likely driven by GFI1 upregulation.
Collapse
Affiliation(s)
- Jingyue Ma
- Department of AnesthesiologyThe First Affiliated Hospital of Chongqing Medical UniversityChongqing400016China
| | - Yichan Ao
- Department of AnesthesiologyThe First Affiliated Hospital of Chongqing Medical UniversityChongqing400016China
| | - Zhen Yue
- Department of AnesthesiologyXinjiang Uygur Autonomous Region Changji People's HospitalChangji831100China
| | - Zhiqiao Wang
- Department of AnesthesiologyThe First Affiliated Hospital of Chongqing Medical UniversityChongqing400016China
| | - Xiangyu Hou
- Department of AnesthesiologyThe First Affiliated Hospital of Chongqing Medical UniversityChongqing400016China
| | - Hongbin Li
- Department of AnesthesiologyThe First Affiliated Hospital of Chongqing Medical UniversityChongqing400016China
| | - Hanbing Wang
- Department of AnesthesiologyThe First Affiliated Hospital of Chongqing Medical UniversityChongqing400016China
| | - Siqing Luo
- Department of AnesthesiologyThe First Affiliated Hospital of Chongqing Medical UniversityChongqing400016China
| | - Jianyu He
- Department of AnesthesiologyThe First Affiliated Hospital of Chongqing Medical UniversityChongqing400016China
| | - Zikun Duan
- Department of AnesthesiologyThe First Affiliated Hospital of Chongqing Medical UniversityChongqing400016China
| | - Ling Liu
- Department of AnesthesiologyThe First Affiliated Hospital of Chongqing Medical UniversityChongqing400016China
| | - Ke Wei
- Department of AnesthesiologyThe First Affiliated Hospital of Chongqing Medical UniversityChongqing400016China
| |
Collapse
|
20
|
Zhao X, Chen C, Qiu H, Liu J, Shao N, Guo M, Jiang Y, Zhao J, Xu L. The landscape of ATF3 in tumors: Metabolism, expression regulation, therapy approach, and open concerns. Pharmacol Res 2025; 214:107666. [PMID: 39978658 DOI: 10.1016/j.phrs.2025.107666] [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: 09/30/2024] [Revised: 02/09/2025] [Accepted: 02/17/2025] [Indexed: 02/22/2025]
Abstract
Cellular stress response is a pivotal process in tumor development and therapy. Activating transcription factor 3 (ATF3), a representative stress-responsive protein, plays pleiotropic roles in various biological processes. Over the past decade, studies have described not only the general role of ATF3 in tumor metabolism but also the complexity of ATF3 expression regulation and its associated modifications, including phosphorylation, ubiquitination, SUMOylation, and NEDDylation. Interestingly, beyond being a transcription factor, ATF3 can act as a modifier to control the ubiquitination of target molecules, such as p53, to exert its function in tumors. These advances in uncovering ATF3 biological function have yielded new insights into the cellular stress response during tumor development and will be instrumental in developing novel interventions. In this review, we update the role of ATF3 as a nexus in amino acid metabolism, lipid metabolism, glycometabolism, and other metabolic pathways in tumors; delineate the underlying mechanisms involving DNA level regulation, epigenetic regulation, and post-translational modifications of ATF3; and summarize the progression of tumor mono/combination therapies related to ATF3. In particular, we discuss the challenges that need to be addressed to provide a new conceptual framework for further understanding the potential therapeutic value of ATF3 in ongoing clinical trials.
Collapse
Affiliation(s)
- Xu Zhao
- Medical College, Guizhou University, Guiyang, Guizhou Province 550025, China; Key Laboratory for Cancer Prevention and Treatment of Guizhou Province, Zunyi, Guizhou 563000, China; Department of Immunology, Zunyi Medical University, Guizhou 563000, China
| | - Chao Chen
- Medical College, Guizhou University, Guiyang, Guizhou Province 550025, China; Key Laboratory for Cancer Prevention and Treatment of Guizhou Province, Zunyi, Guizhou 563000, China; Department of Immunology, Zunyi Medical University, Guizhou 563000, China
| | - Hui Qiu
- Key Laboratory for Cancer Prevention and Treatment of Guizhou Province, Zunyi, Guizhou 563000, China; Department of Immunology, Zunyi Medical University, Guizhou 563000, China
| | - Jing Liu
- Key Laboratory for Cancer Prevention and Treatment of Guizhou Province, Zunyi, Guizhou 563000, China; Department of Immunology, Zunyi Medical University, Guizhou 563000, China
| | - Nan Shao
- Key Laboratory for Cancer Prevention and Treatment of Guizhou Province, Zunyi, Guizhou 563000, China; Department of Immunology, Zunyi Medical University, Guizhou 563000, China
| | - Mengmeng Guo
- Key Laboratory for Cancer Prevention and Treatment of Guizhou Province, Zunyi, Guizhou 563000, China; Department of Immunology, Zunyi Medical University, Guizhou 563000, China
| | - Yuanye Jiang
- Department of Gastroenterology, Putuo hospital, Shanghai University of Tradtional Chinese Medicine, Shanghai 200062, China.
| | - Juanjuan Zhao
- Key Laboratory for Cancer Prevention and Treatment of Guizhou Province, Zunyi, Guizhou 563000, China; Department of Immunology, Zunyi Medical University, Guizhou 563000, China.
| | - Lin Xu
- Medical College, Guizhou University, Guiyang, Guizhou Province 550025, China; Key Laboratory for Cancer Prevention and Treatment of Guizhou Province, Zunyi, Guizhou 563000, China; Department of Immunology, Zunyi Medical University, Guizhou 563000, China; Innovation Center for Tissue Damage Repair, Ministry of Education, Zunyi, Guizhou 563000, China.
| |
Collapse
|
21
|
Honecker B, Bärreiter VA, Höhn K, Horváth B, Harant K, Metwally NG, Marggraff C, Anders J, Leyk S, Martínez-Tauler MDP, Bea A, Hansen C, Fehling H, Lütkemeyer M, Lorenzen S, Franzenburg S, Lotter H, Bruchhaus I. Entamoeba histolytica extracellular vesicles drive pro-inflammatory monocyte signaling. PLoS Negl Trop Dis 2025; 19:e0012997. [PMID: 40208874 PMCID: PMC12052212 DOI: 10.1371/journal.pntd.0012997] [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: 01/08/2025] [Revised: 05/05/2025] [Accepted: 03/19/2025] [Indexed: 04/12/2025] Open
Abstract
The parasitic protozoan Entamoeba histolytica secretes extracellular vesicles (EVs), but so far little is known about their function in the interaction with the host immune system. Infection with E. histolytica trophozoites can lead to formation of amebic liver abscesses (ALAs), in which pro-inflammatory immune responses of Ly6Chi monocytes contribute to liver damage. Men exhibit a more severe pathology as the result of higher monocyte recruitment and a stronger immune response. To investigate the role of EVs and pathogenicity in the host immune response, we studied the effect of EVs secreted by low pathogenic EhA1 and highly pathogenic EhB2 amebae on monocytes. Size and quantity of isolated EVs from both clones were similar. However, they differed in their proteome and miRNA cargo, providing insight into factors potentially involved in amebic pathogenicity. In addition, EVs were enriched in proteins with signaling peptides compared with the total protein content of trophozoites. Exposure to EVs from both clones induced monocyte activation and a pro-inflammatory immune response as evidenced by increased surface presentation of the activation marker CD38 and upregulated gene expression of key signaling pathways (including NF-κB, IL-17 and TNF signaling). The release of pro-inflammatory cytokines was increased in EV-stimulated monocytes and more so in male- than in female-derived cells. While EhA1 EV stimulation caused elevated myeloperoxidase (MPO) release by both monocytes and neutrophils, EhB2 EV stimulation did not, indicating the protective role of MPO during amebiasis. Collectively, our results suggest that parasite-released EVs contribute to the male-biased immunopathology mediated by pro-inflammatory monocytes during ALA formation.
Collapse
Affiliation(s)
- Barbara Honecker
- RG Molecular Infection Immunology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- RG Host-Parasite Interaction, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Valentin A. Bärreiter
- RG Molecular Infection Immunology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- Institute for Infection Research and Vaccine Development, Center for Internal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department for Clinical Immunology of Infectious Diseases, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| | - Katharina Höhn
- Cellular Parasitology Department, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Balázs Horváth
- Arbovirus and Entomology Department, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Karel Harant
- Laboratory of Mass Spectrometry, BIOCEV, Faculty of Science, Charles University in Prague, Prague, Czech Republic
| | - Nahla Galal Metwally
- RG Host-Parasite Interaction, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Claudia Marggraff
- RG Molecular Infection Immunology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Juliett Anders
- RG Host-Parasite Interaction, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Stephanie Leyk
- RG Protozoa Immunology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Maria del Pilar Martínez-Tauler
- RG Host-Parasite Interaction, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- Division of Innate Immunity, Research Center Borstel, Leibniz Lung Center (Airway Research Center North), German Centre for Lung Research, Borstel, Germany
| | - Annika Bea
- RG Molecular Infection Immunology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Charlotte Hansen
- RG Molecular Infection Immunology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Helena Fehling
- RG Molecular Infection Immunology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Melanie Lütkemeyer
- RG Molecular Infection Immunology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Stephan Lorenzen
- Department of Infection Epidemiology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Sören Franzenburg
- Institute of Clinical Molecular Biology, University of Kiel, Kiel, Germany
| | - Hanna Lotter
- RG Molecular Infection Immunology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Iris Bruchhaus
- RG Host-Parasite Interaction, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- Department of Biology, University of Hamburg, Hamburg, Germany
| |
Collapse
|
22
|
O'Carroll SM, O'Neill LAJ. Recycling dead bacteria to fuel macrophage immunometabolism. Nat Immunol 2025; 26:529-530. [PMID: 40108422 DOI: 10.1038/s41590-025-02117-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Affiliation(s)
- Shane M O'Carroll
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland.
| | - Luke A J O'Neill
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland.
| |
Collapse
|
23
|
Han Z, Shen Y, Yan Y, Bin P, Zhang M, Gan Z. Metabolic reprogramming shapes post-translational modification in macrophages. Mol Aspects Med 2025; 102:101338. [PMID: 39977975 DOI: 10.1016/j.mam.2025.101338] [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: 12/10/2024] [Accepted: 01/11/2025] [Indexed: 02/22/2025]
Abstract
Polarized macrophages undergo metabolic reprogramming, as well as extensive epigenetic and post-translational modifications (PTMs) switch. Metabolic remodeling and dynamic changes of PTMs lead to timely macrophage response to infection or antigenic stimulation, as well as its transition from a pro-inflammatory to a reparative phenotype. The transformation of metabolites in the microenvironment also determines the PTMs of macrophages. Here we reviewed the current understanding of the altered metabolites of glucose, lipids and amino acids in macrophages shape signaling and metabolism pathway during macrophage polarization via PTMs, and how these metabolites in some macrophage-associated diseases affect disease progression by shaping macrophage PTMs.
Collapse
Affiliation(s)
- Ziyi Han
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Yinhao Shen
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Yuqi Yan
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, Zhejiang, China
| | - Peng Bin
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Meimei Zhang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China.
| | - Zhending Gan
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China; College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, Jiangsu, China.
| |
Collapse
|
24
|
Qu J, Yin L, Qin S, Sun X, Gong X, Li S, Pan X, Jin Y, Cheng Z, Jin S, Wu W. Identification of the Pseudomonas aeruginosa AgtR-CspC-RsaL pathway that controls Las quorum sensing in response to metabolic perturbation and Staphylococcus aureus. PLoS Pathog 2025; 21:e1013054. [PMID: 40198682 PMCID: PMC12051497 DOI: 10.1371/journal.ppat.1013054] [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: 10/17/2024] [Revised: 04/21/2025] [Accepted: 03/19/2025] [Indexed: 04/10/2025] Open
Abstract
Environmental metabolites and metabolic pathways significantly influence bacterial pathogenesis and interspecies competition. We previously discovered that a mutation in the triosephosphate isomerase gene, tpiA, in Pseudomonas aeruginosa led to defective type III secretion and increased susceptibility to aminoglycoside antibiotics. In this study, we found that the tpiA mutation enhances the Las quorum sensing system due to reduced translation of the negative regulator RsaL. Further investigations demonstrated an upregulation of CspC, a CspA family protein that represses rsaL translation. DNA pull-down assay, along with genetic studies, revealed the role of AgtR in regulating cspC transcription. AgtR is known to regulate pyocyanin production in response to N-acetylglucosamine (GlcNAc), contributing to competition against Staphylococcus aureus. We demonstrated that CspC activates the Las quorum sensing system and subsequent pyocyanin production in response to GlcNAc and S. aureus. Overall, our results elucidate the AgtR-CspC-RsaL-LasI pathway that regulates bacterial virulence factors and its role in competition against S. aureus.
Collapse
Affiliation(s)
- Junze Qu
- Department of Microbiology, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, College of Life Sciences, Nankai University, Tianjin, China
| | - Liwen Yin
- Department of Microbiology, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, College of Life Sciences, Nankai University, Tianjin, China
| | - Shanhua Qin
- Department of Microbiology, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, College of Life Sciences, Nankai University, Tianjin, China
| | - Xiaomeng Sun
- Department of Microbiology, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, College of Life Sciences, Nankai University, Tianjin, China
| | - Xuetao Gong
- Department of Microbiology, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, College of Life Sciences, Nankai University, Tianjin, China
| | - Shouyi Li
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, National Key laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key laboratory of Digestive Cancer, Tianjin’s Clinical Research Center for Cancer, Tianjin, People’s Republic of China
| | - Xiaolei Pan
- Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Yongxin Jin
- Department of Microbiology, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, College of Life Sciences, Nankai University, Tianjin, China
| | - Zhihui Cheng
- Department of Microbiology, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, College of Life Sciences, Nankai University, Tianjin, China
| | - Shouguang Jin
- Department of Microbiology, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, College of Life Sciences, Nankai University, Tianjin, China
| | - Weihui Wu
- Department of Microbiology, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, College of Life Sciences, Nankai University, Tianjin, China
| |
Collapse
|
25
|
Yang Z, Zhang L, Ottavi S, Geri JB, Perkowski A, Jiang X, Pfau D, Bryk R, Aubé J, Zimmerman M, Dartois V, Nathan C. ACOD1-mediated lysosomal membrane permeabilization contributes to Mycobacterium tuberculosis-induced macrophage death. Proc Natl Acad Sci U S A 2025; 122:e2425309122. [PMID: 40100622 PMCID: PMC11962489 DOI: 10.1073/pnas.2425309122] [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: 12/03/2024] [Accepted: 02/18/2025] [Indexed: 03/20/2025] Open
Abstract
Mycobacterium tuberculosis (Mtb) primarily infects macrophages. In vitro without antibiotics, wild-type Mtb hastens death of the macrophages, but the processes leading to rapid cell death are not well understood. Our earlier work indicated that the death of Mtb-infected mouse macrophages in vitro is markedly exacerbated by induction of interferon-β (IFN-β) [L. Zhang et al., J. Exp. Med. 18, e20200887 (2021)]. Here, we identified a key downstream response to IFN-β in the context of Mtb infection as the massive induction of cis-aconitate decarboxylase (ACOD1), not only in its canonical subcellular localization in mitochondria but also in the cytosol, where it bound to the lysosome-stabilizing protein HSP70. ACOD1's product, itaconate, protected Mtb-infected macrophages. However, the contrasting and predominant effect of high-level ACOD1 expression was to act in a noncatalytic manner to promote HSP70's degradation, leading to lysosomal membrane permeabilization (LMP). Mtb-induced macrophage death was markedly diminished by inhibitors of cysteine proteases, consistent with lysosome-mediated cell death. Neither ACOD1 inhibitors nor cysteine protease inhibitors are suitable for potential host-directed therapy (HDT) of tuberculosis. Instead, this work directs attention to how ACOD1 acts nonenzymatically to promote the degradation of HSP70.
Collapse
Affiliation(s)
- Ziwei Yang
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY10065
| | - Li Zhang
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong518055, China
| | - Samantha Ottavi
- Division of Chemical Biology and Medicinal Chemistry, University of North Carolina Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Jacob B. Geri
- Department of Pharmacology, Weill Cornell Medicine, New York, NY10065
| | - Andrew Perkowski
- Division of Chemical Biology and Medicinal Chemistry, University of North Carolina Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Xiuju Jiang
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY10065
| | - Daniel Pfau
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY10065
| | - Ruslana Bryk
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY10065
| | - Jeffrey Aubé
- Division of Chemical Biology and Medicinal Chemistry, University of North Carolina Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Matthew Zimmerman
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ07110
| | - Véronique Dartois
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ07110
| | - Carl Nathan
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY10065
| |
Collapse
|
26
|
Dimitriew W, Schuster S. Dynamic optimization elucidates higher-level pathogenicity strategies of Pseudomonas aeruginosa. MICROLIFE 2025; 6:uqaf005. [PMID: 40182079 PMCID: PMC11967335 DOI: 10.1093/femsml/uqaf005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 02/20/2025] [Accepted: 03/03/2025] [Indexed: 04/05/2025]
Abstract
Multiple dangerous pathogens from the World Health Organization's priority list possess a plethora of virulence components, including the ability to survive inside macrophages. Often, the pathogens rely on a multi-layered defence strategy in order to defend themselves against the immune system. Here, a minimal model is proposed to study such a strategy. By way of example, we consider the interaction between Pseudomonas aeruginosa and the human host, in which the host and the pathogen counter each other in a back-and-forth interaction. In particular, the pathogen attacks the host, macrophages of the host engulf the pathogen and reduce its access to glucose, the pathogen activates the glyoxylate shunt, which is started by the enzyme isocitrate lyase (Icl), the host inhibits it by itaconic acid, and the pathogen metabolizes itaconic acid using the enzyme succinyl-CoA:itaconate CoA transferase (Ict). The flux through the glyoxylate shunt allows the pathogen to avoid carbon loss and oxidative stress. These functions are of utmost importance inside a phagolysosome. Therefore, the pathogen needs to allocate its limited protein resource between the enzymes Icl and Ict in order to maximize the time integral of a flux through the enzyme Icl. We use both random search and dynamic optimization to identify the enzyme Ict as a cost-effective means of counter-counter-counter-defence and as a possible drug target during the early phase of infection.
Collapse
Affiliation(s)
- Wassili Dimitriew
- Department of Bioinformatics, Friedrich Schiller University of Jena, 07743 Jena, Germany
| | - Stefan Schuster
- Department of Bioinformatics, Friedrich Schiller University of Jena, 07743 Jena, Germany
| |
Collapse
|
27
|
Zhu J, Jin Z, Wang J, Wu Z, Xu T, Tong G, Shen E, Fan J, Jiang C, Wang J, Li X, Cong W, Lin L. FGF21 ameliorates septic liver injury by restraining proinflammatory macrophages activation through the autophagy/HIF-1α axis. J Adv Res 2025; 69:477-494. [PMID: 38599281 PMCID: PMC11954821 DOI: 10.1016/j.jare.2024.04.004] [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: 02/02/2024] [Revised: 03/26/2024] [Accepted: 04/07/2024] [Indexed: 04/12/2024] Open
Abstract
INTRODUCTION Sepsis, a systemic immune syndrome caused by severe trauma or infection, poses a substantial threat to the health of patients worldwide. The progression of sepsis is heavily influenced by septic liver injury, which is triggered by infection and cytokine storms, and has a significant impact on the tolerance and prognosis of septic patients. The objective of our study is to elucidate the biological role and molecular mechanism of fibroblast growth factor 21 (FGF21) in the process of sepsis. OBJECTIVES This study was undertaken in an attempt to elucidate the function and molecular mechanism of FGF21 in therapy of sepsis. METHODS Serum concentrations of FGF21 were measured in sepsis patients and septic mice. Liver injury was compared between mice FGF21 knockout (KO) mice and wildtype (WT) mice. To assess the therapeutic potential, recombinant human FGF21 was administered to septic mice. Furthermore, the molecular mechanism of FGF21 was investigated in mice with myeloid-cell specific HIF-1α overexpression mice (LyzM-CreDIO-HIF-1α) and myeloid-cell specific Atg7 knockout mice (Atg7△mye). RESULTS Serum level of FGF21 was significantly increased in sepsis patients and septic mice. Through the use of recombinant human FGF21 (rhFGF21) and FGF21 KO mice, we found that FGF21 mitigated septic liver injury by inhibiting the initiation and propagation of inflammation. Treatment with rhFGF21 effectively suppressed the activation of proinflammatory macrophages by promoting macroautophagy/autophagy degradation of hypoxia-inducible factor-1α (HIF-1α). Importantly, the therapeutic effect of rhFGF21 against septic liver injury was nullified in LyzM-CreDIO-HIF-1α mice and Atg7△mye mice. CONCLUSIONS Our findings demonstrate that FGF21 considerably suppresses inflammation upon septic liver injury through the autophagy/ HIF-1α axis.
Collapse
Affiliation(s)
- Junjie Zhu
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, PR China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, PR China
| | - Zhouxiang Jin
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, PR China
| | - Jie Wang
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, PR China
| | - Zhaohang Wu
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, PR China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, PR China
| | - Tianpeng Xu
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, PR China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, PR China
| | - Gaozan Tong
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, PR China
| | - Enzhao Shen
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, PR China
| | - Junfu Fan
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, PR China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, PR China
| | - Chunhui Jiang
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, PR China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, PR China
| | - Jiaqi Wang
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, PR China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, PR China
| | - Xiaokun Li
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, PR China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, PR China; Department of Hepatobiliary Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, PR China
| | - Weitao Cong
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, PR China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, PR China; Haihe Laboratory of Cell Ecosystem, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, PR China
| | - Li Lin
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, PR China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, PR China.
| |
Collapse
|
28
|
Li J, Guo C, Yang X, Xie W, Mi W, Hua C, Tang C, Wang H. Effects of natural products on macrophage immunometabolism: A new frontier in the treatment of metabolic diseases. Pharmacol Res 2025; 213:107634. [PMID: 39889866 DOI: 10.1016/j.phrs.2025.107634] [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: 10/31/2024] [Revised: 01/20/2025] [Accepted: 01/27/2025] [Indexed: 02/03/2025]
Abstract
Immunometabolic variations in macrophages critically influence their differentiation into pro-inflammatory or anti-inflammatory phenotypes, thereby contributing to immune homeostasis, defense against infection, and tissue repair. Dysregulation of macrophage immunometabolism has been closely implicated in several metabolic diseases, including obesity, type 2 diabetes mellitus (T2DM), non-alcoholic fatty liver disease (NAFLD), hypertension, atherosclerosis, and gout, which positions macrophages as potential therapeutic targets. Recently, several natural products that target macrophage metabolic pathways have shown significant efficacy in managing metabolic diseases; however, a systematic review of these findings has yet to be conducted. This study consolidates natural products with immunoregulatory properties, including flavonoids, phenols, terpenoids, and naphthoquinones, which can alleviate chronic inflammation associated with metabolic disorders by modulating macrophage metabolic pathways, such as aerobic glycolysis, oxidative phosphorylation (OXPHOS), and fatty acid oxidation (FAO). This review aims to elucidate the metabolic regulation of the immune system, analyze metabolic alterations in macrophage associated with metabolic diseases, and summarize the beneficial roles of natural products in immunometabolism, providing novel insights for the prevention and therapeutic management of metabolic diseases.
Collapse
Affiliation(s)
- Jiani Li
- Beijing Academy of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Chen Guo
- Beijing Academy of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Xiaofei Yang
- Beijing University of Chinese Medicine, Beijing 100029, China
| | - Weinan Xie
- Beijing University of Chinese Medicine, Beijing 100029, China
| | - Wenjing Mi
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Chenglong Hua
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Cheng Tang
- Beijing Academy of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China.
| | - Han Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China.
| |
Collapse
|
29
|
Phair IR, Sovakova M, Alqurashi N, Nisr RB, McNeilly AD, Lamont D, Rena G. In-depth proteomic profiling identifies potentiation of the LPS response by 7-ketocholesterol. JOURNAL OF MOLECULAR AND CELLULAR CARDIOLOGY PLUS 2025; 11:100285. [PMID: 39991505 PMCID: PMC11847031 DOI: 10.1016/j.jmccpl.2025.100285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 01/23/2025] [Accepted: 01/28/2025] [Indexed: 02/25/2025]
Abstract
In patients with stable coronary artery disease, plasma levels of 7-ketocholesterol (7-KC), found at high levels in atherosclerotic lesions, predict risk of incident heart failure dose dependently, potentially contributing to disease aetiology. Previous studies demonstrated that 7-KC can elicit effects on macrophage function; however, effects of 7-KC on the macrophage proteome have not been studied systematically. Here we used quantitative mass spectrometry to establish the effect of 7-KC on the mouse macrophage proteome. 7-KC independently mediated dynamic changes, including on atherogenic/M1 markers, cholesterol metabolism, biosynthesis and transport, as well as nutrient transport more broadly. These changes were however insufficient alone to drive changes in cytokine and chemokine secretion. Rather, they prime the macrophage, potentiating LPS-stimulated TNF alpha secretion and key pro-inflammatory enzymes. Our results indicate that 7-KC has independent metabolic effects on the macrophage; however, effects on the immune system are primarily due to the changes in metabolism priming the response to an inflammatory stimulus. Earlier findings from CANTOS and the recent FDA approval of colchicine highlight that inflammation is a viable target for cardiovascular disease; however, it is currrently unclear which will be the best anti-inflammatory targets to pursue in the future. In this context, our findings suggest that drugs targeting atherogenic markers induced by 7-KC might be well tolerated, as they will not necessarily be expected to be immunosuppressive.
Collapse
Affiliation(s)
- Iain R. Phair
- Cellular and Systems Medicine, School of Medicine, University of Dundee, Dundee DD1 9SY, Scotland, United Kingdom
| | - Magdalena Sovakova
- Cellular and Systems Medicine, School of Medicine, University of Dundee, Dundee DD1 9SY, Scotland, United Kingdom
| | - Noor Alqurashi
- Cellular and Systems Medicine, School of Medicine, University of Dundee, Dundee DD1 9SY, Scotland, United Kingdom
| | - Raid B. Nisr
- Cellular and Systems Medicine, School of Medicine, University of Dundee, Dundee DD1 9SY, Scotland, United Kingdom
| | - Alison D. McNeilly
- Cellular and Systems Medicine, School of Medicine, University of Dundee, Dundee DD1 9SY, Scotland, United Kingdom
| | - Douglas Lamont
- Centre for Advanced Scientific Technologies, School of Life Sciences, University of Dundee, Dundee DD1 5EH, Scotland, United Kingdom
| | - Graham Rena
- Cellular and Systems Medicine, School of Medicine, University of Dundee, Dundee DD1 9SY, Scotland, United Kingdom
| |
Collapse
|
30
|
Liu YJ, Sulc J, Auwerx J. Mitochondrial genetics, signalling and stress responses. Nat Cell Biol 2025; 27:393-407. [PMID: 40065146 DOI: 10.1038/s41556-025-01625-w] [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: 11/28/2023] [Accepted: 01/22/2025] [Indexed: 03/15/2025]
Abstract
Mitochondria are multifaceted organelles with crucial roles in energy generation, cellular signalling and a range of synthesis pathways. The study of mitochondrial biology is complicated by its own small genome, which is matrilineally inherited and not subject to recombination, and present in multiple, possibly different, copies. Recent methodological developments have enabled the analysis of mitochondrial DNA (mtDNA) in large-scale cohorts and highlight the far-reaching impact of mitochondrial genetic variation. Genome-editing techniques have been adapted to target mtDNA, further propelling the functional analysis of mitochondrial genes. Mitochondria are finely tuned signalling hubs, a concept that has been expanded by advances in methodologies for studying the function of mitochondrial proteins and protein complexes. Mitochondrial respiratory complexes are of dual genetic origin, requiring close coordination between mitochondrial and nuclear gene-expression systems (transcription and translation) for proper assembly and function, and recent findings highlight the importance of the mitochondria in this bidirectional signalling.
Collapse
Affiliation(s)
- Yasmine J Liu
- Laboratory of Integrative Systems Physiology, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Jonathan Sulc
- Laboratory of Integrative Systems Physiology, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Johan Auwerx
- Laboratory of Integrative Systems Physiology, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.
| |
Collapse
|
31
|
Yu Q, Mei H, Gu Q, Zeng R, Li Y, Zhang J, Gao C, Fang H, Qu J, Liu J. OLFML3 Promotes IRG1 Mitochondrial Localization and Modulates Mitochondrial Function in Macrophages. Int J Biol Sci 2025; 21:2275-2295. [PMID: 40083707 PMCID: PMC11900800 DOI: 10.7150/ijbs.103859] [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: 09/19/2024] [Accepted: 02/15/2025] [Indexed: 03/16/2025] Open
Abstract
Olfactomedin-like protein 3 (OLFML3), belonging to olfactomedin (OLF) protein family, has poorly defined functions. Recent studies have reported the functions of OLFML3 in anti-viral immunity and tumorigenesis. In this study, we investigated the roles of OLFML3 in macrophages. In LPS- or Pseudomonas aeruginosa-induced acute lung injury (ALI) mouse model, OLFML3 depletion exacerbated inflammatory response, leading to reduced survival. OLFML3 achieved the in vivo activity by regulating macrophage phagocytosis and migration. Mass spectrometry analysis revealed immunoresponsive gene 1 (IRG1) as an OLFML3-interacting protein. IRG1 is a mitochondrial decarboxylase that catalyzes the conversion of cis-aconitate to itaconate, a myeloid-borne mitochondrial metabolite with immunomodulatory activities. Further investigation showed that OLFML3 could prevent LPS-induced mitochondrial dysfunction in macrophages by maintaining the homeostasis of mitochondrial membrane potential (MMP), mitochondrial reactive oxygen species (mtROS) and itaconate-related metabolites. In-depth protein-protein interaction studies showed that OLFML3 could promote IRG1 mitochondrial localization via a mitochondrial transport protein, apoptosis inducing factor mitochondria associated 1 (AIFM1). In summary, our study showed that OLFML3 could facilitate IRG1 mitochondrial localization and prevent LPS-induced mitochondrial dysfunction in macrophages.
Collapse
Affiliation(s)
- Qijun Yu
- Department of Pulmonary and Critical Care Medicine, Ruijin Hospital, Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; and Shanghai Key Laboratory of Emergency Prevention, Diagnosis and Treatment of Respiratory Infectious Diseases, Shanghai 200025, China
| | - Hong Mei
- Shanghai Institute for Advanced Immunochemical Studies and School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Qian Gu
- Shanghai Institute for Advanced Immunochemical Studies and School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Ran Zeng
- Department of Pulmonary and Critical Care Medicine, Ruijin Hospital, Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; and Shanghai Key Laboratory of Emergency Prevention, Diagnosis and Treatment of Respiratory Infectious Diseases, Shanghai 200025, China
| | - Yanan Li
- Department of Pulmonary and Critical Care Medicine, Ruijin Hospital, Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; and Shanghai Key Laboratory of Emergency Prevention, Diagnosis and Treatment of Respiratory Infectious Diseases, Shanghai 200025, China
| | - Junjie Zhang
- Shanghai Institute for Advanced Immunochemical Studies and School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Chenxu Gao
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Hai Fang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jieming Qu
- Department of Pulmonary and Critical Care Medicine, Ruijin Hospital, Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; and Shanghai Key Laboratory of Emergency Prevention, Diagnosis and Treatment of Respiratory Infectious Diseases, Shanghai 200025, China
| | - Jia Liu
- Shanghai Institute for Advanced Immunochemical Studies and School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
- Guangzhou Laboratory, Guangzhou International Bio Island, No. 9 XingDaoHuanBei Road, Guangzhou, 510005, Guangdong, China
- Shanghai Clinical Research and Trial Center, Shanghai, 201210, China
| |
Collapse
|
32
|
Li Y, Li Y, Li P, Yang L, Li H. 4-Octyl Itaconate Attenuates Postmenopausal Osteoporosis by Inhibiting Ferroptosis and Enhancing Osteogenesis via the Nrf2 Pathway. Inflammation 2025:10.1007/s10753-025-02268-7. [PMID: 39984770 DOI: 10.1007/s10753-025-02268-7] [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: 01/24/2025] [Accepted: 02/10/2025] [Indexed: 02/23/2025]
Abstract
Bone marrow mesenchymal stem cells (BMSCs) play an important role in bone metabolism and tissue repair, and their ability to differentiate into osteoblasts is crucial in the treatment of bone diseases such as postmenopausal osteoporosis (PMOP). However, the function of BMSCs may be affected by ferroptosis. Ferroptosis is a cell death mode characterized by excess Fe2+ and lipid peroxidation, which significantly affects the survival rate and differentiation ability of BMSCs. This study investigated the effect of exogenous itaconate derivative 4-octyl itaconate (4-OI) on Erastin-induced BMSCs ferroptosis. The results showed that 4-OI significantly inhibited Erastin-induced BMSCs ferroptosis by activating the nuclear factor erythroid 2-related factor 2 (Nrf2) signaling pathway, reduced reactive oxygen species levels and oxidative damage, and restored antioxidant capacity. At the same time, 4-OI promoted the osteogenic differentiation of BMSCs. Further experiments showed that Nrf2-IN-1, an inhibitor of the Nrf2 pathway, could reverse the protective effect of 4-OI. In vivo, 4-OI was shown to reduce bone loss in ovariectomized (OVX) mice, as assessed by Micro-CT analysis. Immunofluorescence staining further revealed increased GPX4 and Nrf2 expression in vertebral tissues following 4-OI treatment. These results indicate that 4-OI improves ferroptosis of BMSCs and enhances osteogenic differentiation ability by activating the Nrf2 pathway, providing new research ideas and potential targets for the treatment of PMOP.
Collapse
Affiliation(s)
- You Li
- Department of Orthopedics, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, 366 Taihu Road, Taizhou, 225300, China
| | - Yang Li
- Department of Orthopedics, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, 366 Taihu Road, Taizhou, 225300, China
| | - Pengfei Li
- School of Postgraduate, Nanjing University of Chinese Medicine, Nanjing, China
| | - Lei Yang
- Department of Orthopedics, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, 366 Taihu Road, Taizhou, 225300, China.
| | - Haijun Li
- Department of Orthopedics, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, 366 Taihu Road, Taizhou, 225300, China.
- School of Postgraduate, Nanjing University of Chinese Medicine, Nanjing, China.
| |
Collapse
|
33
|
Rykova EY, Klimontov VV, Shmakova E, Korbut AI, Merkulova TI, Kzhyshkowska J. Anti-Inflammatory Effects of SGLT2 Inhibitors: Focus on Macrophages. Int J Mol Sci 2025; 26:1670. [PMID: 40004134 PMCID: PMC11854991 DOI: 10.3390/ijms26041670] [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: 11/05/2024] [Revised: 02/04/2025] [Accepted: 02/06/2025] [Indexed: 02/21/2025] Open
Abstract
A growing body of evidence indicates that nonglycemic effects of sodium-glucose cotransporter 2 (SGLT2) inhibitors play an important role in the protective effects of these drugs in diabetes, chronic kidney disease, and heart failure. In recent years, the anti-inflammatory potential of SGLT2 inhibitors has been actively studied. This review summarizes results of clinical and experimental studies on the anti-inflammatory activity of SGLT2 inhibitors, with a special focus on their effects on macrophages, key drivers of metabolic inflammation. In patients with type 2 diabetes, therapy with SGLT2 inhibitors reduces levels of inflammatory mediators. In diabetic and non-diabetic animal models, SGLT2 inhibitors control low-grade inflammation by suppressing inflammatory activation of tissue macrophages, recruitment of monocytes from the bloodstream, and macrophage polarization towards the M1 phenotype. The molecular mechanisms of the effects of SGLT2 inhibitors on macrophages include an attenuation of inflammasome activity and inhibition of the TLR4/NF-κB pathway, as well as modulation of other signaling pathways (AMPK, PI3K/Akt, ERK 1/2-MAPK, and JAKs/STAT). The review discusses the state-of-the-art concepts and prospects of further investigations that are needed to obtain a deeper insight into the mechanisms underlying the effects of SGLT2 inhibitors on the molecular, cellular, and physiological levels.
Collapse
Affiliation(s)
- Elena Y. Rykova
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (IC&G SB RAS), Lavrentjev Prospect 10, 630090 Novosibirsk, Russia; (E.Y.R.); (V.V.K.); (E.S.); (A.I.K.); (T.I.M.)
| | - Vadim V. Klimontov
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (IC&G SB RAS), Lavrentjev Prospect 10, 630090 Novosibirsk, Russia; (E.Y.R.); (V.V.K.); (E.S.); (A.I.K.); (T.I.M.)
- Research Institute of Clinical and Experimental Lymphology, Branch of the Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (RICEL—Branch of IC&G SB RAS), Timakov Str. 2, 630060 Novosibirsk, Russia
| | - Elena Shmakova
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (IC&G SB RAS), Lavrentjev Prospect 10, 630090 Novosibirsk, Russia; (E.Y.R.); (V.V.K.); (E.S.); (A.I.K.); (T.I.M.)
- Laboratory of Translational Cellular and Molecular Biomedicine, National Research Tomsk State University, 634050 Tomsk, Russia
| | - Anton I. Korbut
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (IC&G SB RAS), Lavrentjev Prospect 10, 630090 Novosibirsk, Russia; (E.Y.R.); (V.V.K.); (E.S.); (A.I.K.); (T.I.M.)
- Research Institute of Clinical and Experimental Lymphology, Branch of the Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (RICEL—Branch of IC&G SB RAS), Timakov Str. 2, 630060 Novosibirsk, Russia
| | - Tatyana I. Merkulova
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (IC&G SB RAS), Lavrentjev Prospect 10, 630090 Novosibirsk, Russia; (E.Y.R.); (V.V.K.); (E.S.); (A.I.K.); (T.I.M.)
| | - Julia Kzhyshkowska
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (IC&G SB RAS), Lavrentjev Prospect 10, 630090 Novosibirsk, Russia; (E.Y.R.); (V.V.K.); (E.S.); (A.I.K.); (T.I.M.)
- Laboratory of Translational Cellular and Molecular Biomedicine, National Research Tomsk State University, 634050 Tomsk, Russia
- Institute of Transfusion Medicine and Immunology, Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany
| |
Collapse
|
34
|
Lu S, Gong Y, He P, Qi M, Dong W. 4-octyl Itaconate Attenuates Acute Pancreatitis and Associated Lung Injury by Suppressing Ferroptosis in Mice. Inflammation 2025:10.1007/s10753-025-02256-x. [PMID: 39920558 DOI: 10.1007/s10753-025-02256-x] [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: 10/23/2024] [Revised: 01/14/2025] [Accepted: 01/24/2025] [Indexed: 02/09/2025]
Abstract
Acute pancreatitis (AP) is a common gastrointestinal emergency requiring hospitalization. In recent years, several studies have demonstrated a role for 4-octyl itaconate (4-OI) in anti-inflammatory and oxidative stress injury. However, the potential effects of 4-OI in AP have not been investigated. Caerulein and LPS were used to induce experimental AP models in mice and AR42J cells and then studied by histopathology, biochemical, and molecular analysis. Ferroptosis inhibitor ferrostatin-1 effectively improves pancreatic injury and reduces lipid peroxidation products in experimental AP mice. 4-OI treatment significantly alleviated pancreatic and AP-associated lung injury and inflammation in experimental AP mice by inhibiting ferroptosis. The ferroptosis activator Erastin blocked the protective effect of 4-OI against pancreatic injury in AP, validating that 4-OI alleviates pancreatitis injury through ferroptosis. In vitro experiments further confirmed that 4-OI treatment ameliorated AP-induced pancreatic injury by inhibiting ferroptosis. Our study, for the first time, found that 4-OI ameliorates AP and AP-related lung injury by inhibiting ferroptosis in experimental AP mice, providing a new therapeutic target for alleviating AP.
Collapse
Affiliation(s)
- Shimin Lu
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, China
- Central Laboratory of Renmin Hospital, Wuhan, 430060, Hubei Province, China
| | - Yang Gong
- Central Laboratory of Renmin Hospital, Wuhan, 430060, Hubei Province, China
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, China
| | - Pengzhan He
- Central Laboratory of Renmin Hospital, Wuhan, 430060, Hubei Province, China
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, China
| | - Mingming Qi
- Department of Pathology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, Zhejiang Province, China
| | - Weiguo Dong
- Central Laboratory of Renmin Hospital, Wuhan, 430060, Hubei Province, China.
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, China.
| |
Collapse
|
35
|
Al Akiki Dit Al Mazraani R, Malys N, Maliene V. Itaconate and its derivatives as anti-pathogenic agents. RSC Adv 2025; 15:4408-4420. [PMID: 39931396 PMCID: PMC11808480 DOI: 10.1039/d4ra08298b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 01/24/2025] [Indexed: 02/13/2025] Open
Abstract
Pathogenic microorganisms and viruses cause outbreaks and pandemics that affect millions of people worldwide. Despite recent advances in pharmacology and medicine, the ability of infectious diseases to spread in the modern era is accelerating due to various factors contributing to increased human-to-human and human-animal contacts. With the global rise of drug resistance among pathogens and frequently occurring viral outbreaks, alternative drugs and therapies that specifically inhibit microbial virulence or regulate immune responses are attracting growing interest. The present review focuses on itaconate and its derivatives as potential anti-pathogenic agents. It summarizes the current state of research on itaconate metabolism in bacteria, fungi and mammals. This is followed by a comprehensive review of recent advances studying itaconate and its derivatives as anti-inflammatory, immunoregulatory, antimicrobial and antiviral compounds, along with their mechanisms of action. Finally, the review emphasises the existing challenges and future research directions for the application of itaconate and its derivatives as anti-pathogenic agents.
Collapse
Affiliation(s)
| | - Naglis Malys
- Bioprocess Research Centre, Faculty of Chemical Technology, Kaunas University of Technology Radvilėnų st. 19 Kaunas LT-50254 Lithuania
- Department of Organic Chemistry, Faculty of Chemical Technology, Kaunas University of Technology Radvilėnų st. 19 Kaunas LT-50254 Lithuania
| | - Vida Maliene
- Built Environment and Sustainable Technologies Research Institute, Faculty of Health, Innovation, Technology and Science, Liverpool John Moores University Byrom Street Liverpool L3 3AF UK
| |
Collapse
|
36
|
Woodworth KE, Froom ZSCS, Osborne ND, Rempe CN, Wheeler B, Medd K, Callaghan NI, Qian H, Acharya AP, Charron C, Huyer LD. Development of itaconate polymer microparticles for intracellular regulation of pro-inflammatory macrophage activation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.30.635692. [PMID: 39974988 PMCID: PMC11838496 DOI: 10.1101/2025.01.30.635692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Itaconate (IA) is an endogenous metabolite and a potent regulator of the innate immune system. Its use in immunomodulatory therapies has faced limitations due to inherent challenges in achieving controlled delivery and requirements for high extracellular concentrations to achieve internalization of the highly polar small molecule to achieve its intracellular therapeutic activity. Microparticle (MP)-based delivery strategies are a promising approach for intracellular delivery of small molecule metabolites through macrophage phagocytosis and subsequent intracellular polymer degradation-based delivery. Toward the goal of intracellular delivery of IA, degradable polyester polymer-(poly(itaconate-co-dodecanediol)) based IA polymer microparticles (IA-MPs) were generated using an emulsion method, forming micron-scale (∼ 1.5 µm) degradable microspheres. IA-MPs were characterized with respect to their material properties and IA release kinetics to inform particle fabrication. Treatment of murine bone marrow-derived macrophages with an optimized particle concentration of 0.1 mg/million cells enabled phagocytosis-mediated internalization and low levels of cytotoxicity. Flow cytometry demonstrated IA-MP-specific regulation of IA-sensitive inflammatory targets. Metabolic analyses demonstrated that IA-MP internalization inhibited oxidative metabolism and induced glycolytic reliance, consistent with the established mechanism of IA-associated inhibition of succinate dehydrogenase. This development of IA-based polymer microparticles provides a basis for additional innovative metabolite-based microparticle drug delivery systems for the treatment of inflammatory disease.
Collapse
|
37
|
Zeng L, Wang Y, Huang Y, Yang W, Zhou P, Wan Y, Tao K, Li R. IRG1/itaconate enhances efferocytosis by activating Nrf2-TIM4 signaling pathway to alleviate con A induced autoimmune liver injury. Cell Commun Signal 2025; 23:63. [PMID: 39910615 PMCID: PMC11796036 DOI: 10.1186/s12964-025-02075-5] [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: 09/28/2024] [Accepted: 01/30/2025] [Indexed: 02/07/2025] Open
Abstract
Immune response gene 1 (IRG1) is highly expressed in mitochondria of macrophages in a pro-inflammatory state. IRG1 and its metabolites play important roles in infection, immune-related diseases and tumor progression by exerting resistance of pathogens, attenuating inflammation and producing antioxidant substances through various pathways and mechanisms. IRG1 deficiency aggravates liver injury. Efferocytosis is a vital mechanism for preventing the progression of inflammatory tissue damage. However, the mechanism by how IRG1/itaconate regulates efferocytosis in autoimmune hepatitis has yet to be fully understood. Therefore, we explored the influence of IRG1-/- on efferocytosis and its effects on regulating the nuclear factor erythroid 2-associated factor 2 (Nrf2)-T-cell immunoglobulin domain and mucin domain 4 (TIM4) pathway and autoimmune liver injury. An autoimmune hepatitis model was established by injecting Con A into wild-type and IRG1-/- mice via the tail vein. Liver injury and inflammatory response were assessed. The efferocytosis role of IRG1-/- macrophages and its potential regulatory mechanisms were also analysed. Exogenous 4-octyl itaconate (OI) supplementation promoted the expression of Nrf2 and TIM4 and restored IRG1-/- bone marrow-derived macrophage (BMDM) efferocytosis, whereas inhibition of Nrf2 mediated by ML385 led to impaired efferocytosis of BMDMs, decreased expression of TIM4, and aggravated liver inflammation injury. Additionally, after supplementing Nrf2-/- BMDMs with exogenous OI, we evaluated the changes in its efferocytosis effect, efferocytosis did not change, and the protective effect of OI disappeared. However, when TIM4 was blocked, the efferocytotic effect of BMDMs was attenuated, inflammatory liver injury and oxidative stress were aggravated. OI promoted the transformation of macrophages into M2 macrophages, and this was inhibited when TIM4 was blocked. To our best understanding, this is the initial exploration to show that TIM4, a downstream molecule of the IRG1/itaconate-Nrf2 pathway, regulates macrophage efferocytosis. These findings suggest a new mechanism and potential treatment for promoting the resolution of inflammation and efferocytosis in autoimmune hepatitis.
Collapse
Affiliation(s)
- Liwu Zeng
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022, Hubei Province, China
| | - Yaxin Wang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Yongzhou Huang
- Department of General Surgery, First Affiliated Hospital of Shihezi University, Shihezi, 832000, Xinjiang, China
| | - Wenchang Yang
- Department of Gastroenterology Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Pei Zhou
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022, Hubei Province, China
| | - Yaqi Wan
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022, Hubei Province, China
| | - Kaixiong Tao
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022, Hubei Province, China.
| | - Ruidong Li
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022, Hubei Province, China.
| |
Collapse
|
38
|
Fan Y, Dan W, Wang Y, Ma Z, Jian Y, Liu T, Li M, Wang Z, Wei Y, Liu B, Ding P, Lei Y, Guo C, Zeng J, Yan X, Wei W, Li L. Itaconate transporter SLC13A3 confers immunotherapy resistance via alkylation-mediated stabilization of PD-L1. Cell Metab 2025; 37:514-526.e5. [PMID: 39809284 DOI: 10.1016/j.cmet.2024.11.012] [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: 04/29/2024] [Revised: 08/27/2024] [Accepted: 11/25/2024] [Indexed: 01/16/2025]
Abstract
Itaconate is a metabolite catalyzed by cis-aconitate decarboxylase (ACOD1), which is mainly produced by activated macrophages and secreted into the extracellular environment to exert complex bioactivity. In the tumor microenvironment, itaconate is concentrated and induces an immunosuppressive response. However, whether itaconate can be taken up by tumor cells and its mechanism of action remain largely unclear. Here, we identified solute carrier family 13 member 3 (SLC13A3) as a key protein transporting extracellular itaconate into cells, where it elevates programmed cell death ligand 1 (PD-L1) protein levels and decreases the expression of immunostimulatory molecules, thereby promoting tumor immune evasion. Mechanistically, itaconate alkylates the cysteine 272 residue on PD-L1, antagonizing PD-L1 ubiquitination and degradation. Consequently, SLC13A3 inhibition enhances the efficacy of anti-CTLA-4 (cytotoxic T lymphocyte-associated antigen-4) immunotherapy and improves the overall survival rate in syngeneic mouse tumor models. Collectively, our findings identified SLC13A3 as a key transporter of itaconate and revealed its immunomodulatory role, providing combinatorial strategies to overcome immunotherapy resistance in tumors.
Collapse
Affiliation(s)
- Yizeng Fan
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China; Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi 710061, P.R. China
| | - Weichao Dan
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China; Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi 710061, P.R. China
| | - Yuzhao Wang
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China; Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi 710061, P.R. China
| | - Zhiqiang Ma
- Department of Medical Oncology, Senior Department of Oncology, Chinese PLA General Hospital, The Fifth Medical Center, Beijing 100853, P.R. China
| | - Yanlin Jian
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China; Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi 710061, P.R. China
| | - Tianjie Liu
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China; Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi 710061, P.R. China
| | - Mengxing Li
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China; Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi 710061, P.R. China
| | - Zixi Wang
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China; Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi 710061, P.R. China
| | - Yi Wei
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China; Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi 710061, P.R. China
| | - Bo Liu
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China; Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi 710061, P.R. China
| | - Peng Ding
- Department of Thoracic Surgery, Tangdu Hospital, The Air Force Military Medical University, Xi'an, Shaanxi 710038, P.R. China
| | - Yuzeshi Lei
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China; Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi 710061, P.R. China
| | - Chendong Guo
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China; Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi 710061, P.R. China
| | - Jin Zeng
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China; Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi 710061, P.R. China
| | - Xiaolong Yan
- Department of Thoracic Surgery, Tangdu Hospital, The Air Force Military Medical University, Xi'an, Shaanxi 710038, P.R. China
| | - Wenyi Wei
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| | - Lei Li
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China; Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi 710061, P.R. China.
| |
Collapse
|
39
|
He Z, Pan X, Xie K, Sakao K, Chen J, Komatsu M, Hou DX. The Effects of Fisetin on Gene Expression Profile and Cellular Metabolism in IFN-γ-Stimulated Macrophage Inflammation. Antioxidants (Basel) 2025; 14:182. [PMID: 40002369 PMCID: PMC11852181 DOI: 10.3390/antiox14020182] [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: 11/23/2024] [Revised: 01/22/2025] [Accepted: 02/01/2025] [Indexed: 02/27/2025] Open
Abstract
Although interferon-gamma (IFN-γ) is known as a critical factor in polarizing macrophages into the pro-inflammatory state for immune response, how dietary flavonoids regulate IFN-γ response for anti-inflammation is incompletely elucidated. This study aims to investigate the effect of fisetin, a typical flavonol, on the inhibition of IFN-γ-induced inflammation by RNA sequencing (RNA-Seq) and cellular metabolism analysis. RAW264 macrophages pretreated with fisetin following IFN-γ stimulation were subjected to RNA-Seq to analyze alterations in gene expression. Cellular signaling and transcription were investigated using enrichment analysis, motif analysis, and transcription factor prediction. Cellular metabolic state was assessed by measuring the oxygen consumption rate (OCR) and lactate level to reflect mitochondrial respiration and glycolysis. Alterations in signaling proteins were confirmed by Western blot. The results revealed that fisetin downregulated the IFN-γ-induced expression of pro-inflammatory genes and M1 marker genes such as Cxcl9, Il6, Cd80, Cd86, and Nos2. In cellular metabolism, fisetin upregulated the oxidative phosphorylation (OXPHOS) pathway, restored impaired OCR, and reduced lactate production induced by IFN-γ. Motif analysis suggested that fisetin suppressed the activation of IFN-regulatory factor 1 (IRF1). Western blot data further confirmed that fisetin inhibited the phosphorylation of Jak1, Jak2, and STAT1, and decreased the nuclear accumulation of phosphorylated STAT1 and IRF1 induced by IFN-γ. Taken together, our data revealed that fisetin is a potent flavonoid that attenuates IFN-γ-stimulated murine macrophage inflammation and ameliorates disrupted cellular metabolism with a possible Jak1/2-STAT1-IRF1 pathway.
Collapse
Affiliation(s)
- Ziyu He
- The United Graduate School of Agricultural Sciences, Kagoshima University, Kagoshima 890-0065, Japan; (Z.H.); (K.S.); (M.K.)
| | - Xuchi Pan
- Graduate School of Agriculture, Forestry and Fisheries, Kagoshima University, Kagoshima 890-0065, Japan
| | - Kun Xie
- Hunan Collaborative Innovation Center for Utilization of Botanical Functional Ingredients, College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Kozue Sakao
- The United Graduate School of Agricultural Sciences, Kagoshima University, Kagoshima 890-0065, Japan; (Z.H.); (K.S.); (M.K.)
- Graduate School of Agriculture, Forestry and Fisheries, Kagoshima University, Kagoshima 890-0065, Japan
| | - Jihua Chen
- Department of Nutrition Science and Food Hygiene, Xiangya School of Public Health, Central South University, Changsha 410128, China;
| | - Masaharu Komatsu
- The United Graduate School of Agricultural Sciences, Kagoshima University, Kagoshima 890-0065, Japan; (Z.H.); (K.S.); (M.K.)
- Graduate School of Agriculture, Forestry and Fisheries, Kagoshima University, Kagoshima 890-0065, Japan
| | - De-Xing Hou
- The United Graduate School of Agricultural Sciences, Kagoshima University, Kagoshima 890-0065, Japan; (Z.H.); (K.S.); (M.K.)
- Graduate School of Agriculture, Forestry and Fisheries, Kagoshima University, Kagoshima 890-0065, Japan
| |
Collapse
|
40
|
Luo Y, Jiang LY, Liao ZZ, Wang YY, Wang YD, Xiao XH. Metabolic Regulation of Inflammation: Exploring the Potential Benefits of Itaconate in Autoimmune Disorders. Immunology 2025; 174:189-202. [PMID: 39542834 DOI: 10.1111/imm.13875] [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: 09/08/2024] [Revised: 10/21/2024] [Accepted: 10/22/2024] [Indexed: 11/17/2024] Open
Abstract
Itaconic acid and its metabolites have demonstrated significant therapeutic potential in various immune diseases. Originating from the tricarboxylic acid cycle in immune cells, itaconic acid can modulate immune responses, diminish inflammation, and combat oxidative stress. Recent research has uncovered multiple mechanisms through which itaconic acid exerts its effects, including the inhibition of inflammatory cytokine production, activation of anti-inflammatory pathways, and modulation of immune cell function by regulating cellular metabolism. Cellular actions are influenced by the modulation of metabolic pathways, such as inhibiting succinate dehydrogenase (SDH) activity or glycolysis, activation of nuclear-factor-E2-related factor 2 (Nrf2), boosting cellular defences against oxidative stress, and suppression of immune cell inflammation through the NF-κB pathway. This comprehensive review discusses the initiation, progression, and mechanisms of action of itaconic acid and its metabolites, highlighting their modulatory effects on various immune cell types. Additionally, it examines their involvement in immune disease like rheumatoid arthritis, multiple sclerosis, type 1 diabetes mellitus, and autoimmune hepatitis, offering greater understanding for creating new therapies for these ailments.
Collapse
Affiliation(s)
- Yin Luo
- The First Affiliated Hospital, Department of Metabolism and Endocrinology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Li-Yan Jiang
- The First Affiliated Hospital, Department of Metabolism and Endocrinology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Zhe-Zhen Liao
- The First Affiliated Hospital, Department of Metabolism and Endocrinology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Yuan-Yuan Wang
- The First Affiliated Hospital, Department of Metabolism and Endocrinology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Ya-Di Wang
- The First Affiliated Hospital, Department of Metabolism and Endocrinology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Xin-Hua Xiao
- The First Affiliated Hospital, Department of Metabolism and Endocrinology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| |
Collapse
|
41
|
Zhao N, Yi M, Zhang LJ, Zhang QX, Yang L. 4-Octyl Itaconate Attenuates Neuroinflammation in Experimental Autoimmune Encephalomyelitis Via Regulating Microglia. Inflammation 2025; 48:151-164. [PMID: 38761250 DOI: 10.1007/s10753-024-02050-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 05/02/2024] [Accepted: 05/08/2024] [Indexed: 05/20/2024]
Abstract
Abnormal activation of microglia, the resident macrophages in the central nervous system, plays an important role in the pathogenesis of multiple sclerosis (MS). The immune responsive gene 1(IRG1)/itaconate axis is involved in regulating microglia-mediated neuroinflammation. 4-Octyl itaconate (4-OI), a derivative of itaconate, plays a crucial immunomodulatory role in macrophages. This study investigated the effects and mechanisms of action of 4-OI on experimental autoimmune encephalomyelitis (EAE) and inflammatory BV2 microglia. In an EAE mouse model, clinical evaluation was conducted during the disease course. Hematoxylin and eosin staining was performed to assess inflammatory infiltration and Luxol Fast Blue was used to visualize pathological damage. Quantitative real-time polymerase chain reaction, western blotting and immunofluorescence were used to evaluate inflammatory response and microglial function status in EAE mice. BV2 microglia were used to further investigate the effects and mechanisms of action of 4-OI in vitro. 4-OI significantly alleviated the clinical symptoms of EAE, the inflammatory infiltration, and demyelination; reduced the levels of inflammatory factors; and inhibited the classical activation of microglia in the spinal cord. 4-OI successfully suppressed the classical activation of BV2 microglia and decreased the levels of inflammatory factors by activating the Nrf2/HO-1 signaling pathway. Furthermore, 4-OI downregulated IRG1 expression in both EAE mice and inflammatory BV2 microglia. 4-OI attenuates the microglia-mediated neuroinflammation and has promising therapeutic effects in MS.
Collapse
Affiliation(s)
- Ning Zhao
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Ming Yi
- Department of The Key Laboratory for Human Disease Gene Study of Sichuan Province and the Department of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, 611731, China
- Research Unit for Blindness Prevention of the Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, China
| | - Lin-Jie Zhang
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Qiu-Xia Zhang
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Li Yang
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China.
| |
Collapse
|
42
|
Cummins CL, Goldstein I. New anti-inflammatory mechanism of glucocorticoids uncovered. Trends Endocrinol Metab 2025; 36:99-101. [PMID: 39181729 DOI: 10.1016/j.tem.2024.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 08/08/2024] [Accepted: 08/08/2024] [Indexed: 08/27/2024]
Abstract
Glucocorticoids (GCs) are potent anti-inflammatory drugs. A new study by Auger et al. found that GCs increase itaconate, an anti-inflammatory tricarboxylic acid (TCA) cycle intermediate, by promoting movement of cytosolic pyruvate dehydrogenase (PDH) to mitochondria. Itaconate was sufficient for mediating the anti-inflammatory effects of GCs in mice, overriding the notion that nuclear glucocorticoid receptor (GR) is necessary for inflammation inhibition.
Collapse
Affiliation(s)
- Carolyn L Cummins
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, M5S 3M2, Canada.
| | - Ido Goldstein
- Institute of Biochemistry, Food Science and Nutrition, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, 7610001, Israel
| |
Collapse
|
43
|
Swarnkar G, Naaz M, Mims D, Gupta P, Peterson T, Christopher MJ, Singamaneni S, Mbalaviele G, Abu-Amer Y. IĸBζ as a Central Modulator of Inflammatory Arthritis Pathogenesis. Arthritis Rheumatol 2025; 77:124-139. [PMID: 39279148 PMCID: PMC11785494 DOI: 10.1002/art.42990] [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: 09/17/2023] [Revised: 09/06/2024] [Accepted: 09/10/2024] [Indexed: 09/18/2024]
Abstract
OBJECTIVE Current therapies targeting individual factors in inflammatory arthritis show variable efficacy, often requiring treatment with combinations of drugs, and are associated with undesirable side effects. NF-ĸB is critical for the production and function of most inflammatory cytokines. However, given its essential role in physiologic processes, targeting NF-ĸB is precarious. Hence, identifying pathways downstream of NF-ĸB that selectively govern the expression of inflammatory cytokines in inflammatory arthritis would be advantageous. We have previously identified IĸBζ as a unique inflammatory signature of NF-ĸB that controls the transcription of inflammatory cytokines only under pathologic conditions while sparing physiologic NF-ĸB signals. METHODS We generated mice harboring myeloid, lymphoid, and global deletion of Nfkbiz (the gene encoding IĸBζ). These models were subjected to serum transfer-induced arthritis. Additionally, pharmacologic inhibitors of IĸBζ were injected intraperitonially. Joint swelling, microcomputed tomography, immunohistochemistry, flow cytometry, and cytokine measurements were conducted using synovial tissue samples. RESULTS Global deletion of Nfkbiz or depletion of neutrophils (vastly IĸBζ+ cells) reduced inflammatory synovial cells and increased anti-inflammatory and regenerative synovial cells, plummeted expression of inflammatory factors and ameliorated experimental mouse inflammatory arthritis. Further, expression of immune responsive gene-1, the enzyme responsible for itaconate production, was increased in synovial cells. Accordingly, the itaconate derivative dimethyl itaconate (DI) inhibited IĸBζ-mediated inflammatory factors. Further, in silico screen identified 8-hydroxyquinoline (HQ) as a putative inhibitor of IĸBζ not affecting physiologic NF-ĸB activity. Congruently, systemic administration of either DI or HQ inhibited joint swelling and damage. CONCLUSION Our study positions IĸBζ as an inflammation-specific target for therapeutic consideration in rheumatoid arthritis because its inhibition spares the beneficial functions of NF-ĸB.
Collapse
Affiliation(s)
- Gaurav Swarnkar
- Department of Orthopedic Surgery, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Musarrat Naaz
- Department of Orthopedic Surgery, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Dorothy Mims
- Department of Orthopedic Surgery, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Prashant Gupta
- Department of Mechanical Engineering and Material Science, Washington University in St. Louis, Missouri 63130
| | - Timothy Peterson
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110
- HealthSpan Technologies, Inc. St. Louis, Missouri 63110
- Bioio, Inc. St. Louis, Missouri 63110
| | - Matthew J. Christopher
- Division of Oncology, Cellular Therapy Section, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Srikanth Singamaneni
- Department of Mechanical Engineering and Material Science, Washington University in St. Louis, Missouri 63130
| | - Gabriel Mbalaviele
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Yousef Abu-Amer
- Department of Orthopedic Surgery, Washington University School of Medicine, St. Louis, Missouri 63110
- Department of Cell Biology & Physiology, Washington University School of Medicine, St. Louis, Missouri 63110
- Shriners Hospital for Children, St. Louis, Missouri 63110
| |
Collapse
|
44
|
Wang C, Lv J, Yang M, Fu Y, Wang W, Li X, Yang Z, Lu J. Recent advances in surface functionalization of cardiovascular stents. Bioact Mater 2025; 44:389-410. [PMID: 39539518 PMCID: PMC11558551 DOI: 10.1016/j.bioactmat.2024.10.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 10/21/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
Cardiovascular diseases (CVD) are the leading global threat to human health. The clinical application of vascular stents improved the survival rates and quality of life for patients with cardiovascular diseases. However, despite the benefits stents bring to patients, there are still notable complications such as thrombosis and in-stent restenosis (ISR). Surface modification techniques represent an effective strategy to enhance the clinical efficacy of vascular stents and reduce complications. This paper reviews the development strategies of vascular stents based on surface functional coating technologies aimed at addressing the limitations in clinical application, including the inhibition of intimal hyperplasia, promotion of re-endothelialization. These strategies have improved endothelial repair and inhibited vascular remodeling, thereby promoting vascular healing post-stent implantation. However, the pathological microenvironment of target vessels and the lipid plaques are key pathological factors in the development of atherosclerosis (AS) and impaired vascular repair after percutaneous coronary intervention (PCI). Therefore, restoring normal physiological environment and removing the plaques are also treatment focuses after PCI for promoting vascular repair. Unfortunately, research in this area is limited. This paper reviews the advancements in vascular stents based on surface engineering technologies over the past decade, providing guidance for the development of stents.
Collapse
Affiliation(s)
- Chuanzhe Wang
- Dongguan Key Laboratory of Smart Biomaterials and Regenerative Medicine, The Tenth Affiliated Hospital of Southern Medical University, 523059, Dongguan, Guangdong, China
| | - Jie Lv
- Department of Anesthesiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, 610072, Chengdu, Sichuan, China
| | - Mengyi Yang
- School of Materials Science and Engineering, Key Lab of Advanced Technology for Materials of Education Ministry, Southwest Jiaotong University, 610031, Chengdu, China
| | - Yan Fu
- Dongguan Key Laboratory of Smart Biomaterials and Regenerative Medicine, The Tenth Affiliated Hospital of Southern Medical University, 523059, Dongguan, Guangdong, China
| | - Wenxuan Wang
- School of Materials Science and Engineering, Key Lab of Advanced Technology for Materials of Education Ministry, Southwest Jiaotong University, 610031, Chengdu, China
| | - Xin Li
- Department of Cardiology, Third People's Hospital of Chengdu Affiliated to Southwest Jiaotong University, 610072, Chengdu, Sichuan, China
| | - Zhilu Yang
- Dongguan Key Laboratory of Smart Biomaterials and Regenerative Medicine, The Tenth Affiliated Hospital of Southern Medical University, 523059, Dongguan, Guangdong, China
| | - Jing Lu
- Department of Anesthesiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, 610072, Chengdu, Sichuan, China
| |
Collapse
|
45
|
Zhou P, Yang L, Li H, Zeng L, Zhang Y, Zhong Z, Li R, Yin Y, Tao K, Zhang P. IRG1/Itaconate inhibits hepatic stellate cells ferroptosis and attenuates TAA-induced liver fibrosis by regulating SLC39A14 expression. Int Immunopharmacol 2025; 146:113945. [PMID: 39724735 DOI: 10.1016/j.intimp.2024.113945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 12/13/2024] [Accepted: 12/22/2024] [Indexed: 12/28/2024]
Abstract
This study aimed to elucidate the protective roles of Immune Response Gene-1 (IRG1) and exogenous itaconate in murine models of hepatic fibrosis and to delineate the underlying mechanistic pathways using both wild-type and IRG1-deficient (IRG1-/-) mice. Primary murine stellate cells (mHSC) and bone marrow-derived macrophages (BMDM) were isolated and cocultured. Hepatocellular fibrosis was induced in vitro using Transforming Growth Factor-beta (TGF-β) to evaluate the protective efficacy of IRG1/itaconate. Histopathological damage in the hepatic tissues was assessed using Hematoxylin and Eosin (H&E), Masson's trichrome, and Sirius red staining, followed by hepatic fibrosis scoring. The levels of released inflammatory cytokines were quantified using enzyme-linked immunosorbent assay (ELISA) kits. Immunohistochemistry was used to detect 4-Hydroxynonenal (4-HNE) levels and Perls staining was used to assess ferroptosis. RNA sequencing and gene enrichment analyses were performed to identify implicated molecular entities and signaling pathways. IRG1 and SLC39A14 knockdown and overexpression cell lines were generated. Quantitative real-time PCR (qRT-PCR) and western blotting (WB) were used to measure the mRNA and protein expression levels in hepatic tissues and cells. Kits were used to assess reactive oxygen species (ROS) and malondialdehyde (MDA) levels, and the concentrations of liver enzymes, iron, GSH, and GSSG within hepatic tissues and cells.4-octyl itaconate (4-OI) significantly attenuated the histopathological damage in hepatic tissues, preserved the normal hepatic function, effectively reduced the release of inflammatory cytokines, and mitigated oxidative stress markers such as ROS and MDA in Thioacetamide (TAA)-induced fibrotic mice. Notably, this study is the first to reveal the pivotal role of SLC39A14 in the pathogenesis of hepatic fibrosis in murine models and elucidate how IRG1/itaconate mediates downstream ferroptosis-related signaling pathways by targeting SLC39A14, thereby inhibiting ferroptosis-induced hepatic fibrosis. IRG1/itaconate can alleviate the TAA-induced hepatic fibrosis in mice by regulating the expression of SLC39A14, consequently suppressing hepatic stellate cell ferroptosis.
Collapse
Affiliation(s)
- Pei Zhou
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Lei Yang
- Department of General Surgery, The Second Hospital of Shanxi Medical University, Taiyuan 030001, China
| | - Hang Li
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Liwu Zeng
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yizhuo Zhang
- Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Ziyou Zhong
- Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Ruidong Li
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yuping Yin
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Kaixiong Tao
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Peng Zhang
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
46
|
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
|
47
|
Wu J, Singh K, Shing V, Gupta A, Arenberg BC, Huffstutler RD, Lee DY, Sack MN. Mitochondrial fatty acid oxidation regulates monocytic type I interferon signaling via histone acetylation. SCIENCE ADVANCES 2025; 11:eadq9301. [PMID: 39841826 PMCID: PMC11753372 DOI: 10.1126/sciadv.adq9301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 12/19/2024] [Indexed: 01/24/2025]
Abstract
Although lipid-derived acetyl-coenzyme A (CoA) is a major carbon source for histone acetylation, the contribution of fatty acid β-oxidation (FAO) to this process remains poorly characterized. To investigate this, we generated mitochondrial acetyl-CoA acetyltransferase 1 (ACAT1, distal FAO enzyme) knockout macrophages. 13C-carbon tracing confirmed reduced FA-derived carbon incorporation into histone H3, and RNA sequencing identified diminished interferon-stimulated gene expression in the absence of ACAT1. Chromatin accessibility at the Stat1 locus was diminished in ACAT1-/- cells. Chromatin immunoprecipitation analysis demonstrated reduced acetyl-H3 binding to Stat1 promoter/enhancer regions, and increasing histone acetylation rescued Stat1 expression. Interferon-β release was blunted in ACAT1-/- and recovered by ACAT1 reconstitution. Furthermore, ACAT1-dependent histone acetylation required an intact acetylcarnitine shuttle. Last, obese subjects' monocytes exhibited increased ACAT1 and histone acetylation levels. Thus, our study identifies an intriguing link between FAO-mediated epigenetic control of type I interferon signaling and uncovers a potential mechanistic nexus between obesity and type I interferon signaling.
Collapse
Affiliation(s)
- Jing Wu
- Laboratory of Mitochondrial Biology and Metabolism, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Komudi Singh
- Laboratory of Mitochondrial Biology and Metabolism, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Vivian Shing
- Laboratory of Mitochondrial Biology and Metabolism, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Anand Gupta
- Laboratory of Mitochondrial Biology and Metabolism, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Brett C. Arenberg
- Laboratory of Mitochondrial Biology and Metabolism, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Rebecca D. Huffstutler
- Cardiovascular Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Duck-Yeon Lee
- Biochemistry Core, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Michael N. Sack
- Laboratory of Mitochondrial Biology and Metabolism, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
- Cardiovascular Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
48
|
Zhang SM, Yan G, Lekired A, Zhong D. Genomic basis of schistosome resistance in a molluscan vector of human schistosomiasis. iScience 2025; 28:111520. [PMID: 39758819 PMCID: PMC11699755 DOI: 10.1016/j.isci.2024.111520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 10/15/2024] [Accepted: 11/29/2024] [Indexed: 01/07/2025] Open
Abstract
Freshwater snails are obligate intermediate hosts for the transmission of schistosomiasis, one of the world's most devastating parasitic diseases. To decipher the mechanisms underlying snail resistance to schistosomes, recombinant inbred lines (RILs) were developed from two well-defined homozygous lines (iM line and iBS90) of the snail Biomphalaria glabrata. Whole-genome sequencing (WGS) was used to scan the genomes of 46 individual RIL snails, representing 46 RILs, half of which were resistant or susceptible to Schistosoma mansoni. Genome-wide association study (GWAS) and bin marker-assisted quantitative trait loci (QTLs) analysis, aided by our chromosome-level assembled genome, were conducted. A small genomic region (∼3 Mb) on chromosome 5 was identified as being associated with schistosome resistance, designated the B. glabrata schistosome resistance region 1 (BgSRR1). This study, built on our recently developed genetic and genomic resources, provides valuable insights into anti-schistosome mechanisms and the future development of snail-targeted biocontrol programs for schistosomiasis.
Collapse
Affiliation(s)
- Si-Ming Zhang
- Center for Evolutionary and Theoretical Immunology, Department of Biology, University of New Mexico, Albuquerque, NM 87131, USA
| | - Guiyun Yan
- Program in Public Health, College of Health Sciences, University of California, Irvine, Irvine, CA 92697, USA
| | - Abdelmalek Lekired
- Center for Evolutionary and Theoretical Immunology, Department of Biology, University of New Mexico, Albuquerque, NM 87131, USA
| | - Daibin Zhong
- Program in Public Health, College of Health Sciences, University of California, Irvine, Irvine, CA 92697, USA
| |
Collapse
|
49
|
Switala L, Di L, Colantonio S, Lakshman B, Caceres TW, Reading JJ, Garcia-Buntley SS, Maiseyeu A. The Development and Characterization of Two Monoclonal Antibodies Against the Conjugates and Derivatives of the Immunometabolite Itaconate. ACS OMEGA 2025; 10:1110-1121. [PMID: 39829496 PMCID: PMC11740141 DOI: 10.1021/acsomega.4c08552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 12/04/2024] [Accepted: 12/10/2024] [Indexed: 01/22/2025]
Abstract
We have developed two monoclonal antibodies, CPTC-2MeSC-1 and CPTC-2MeSC-2, against itaconate and its conjugates with sulfhydryl-containing biomolecules such as cysteines. Itaconate is a dicarboxylic acid metabolite that has recently gained much interest for its anti-inflammatory properties in many biological models. We have synthesized an itaconate-cysteine conjugate ITA-Cys designed to mimic in vivo Michael adducts of itaconate. Two monoclonal antibodies against ITA-Cys, CPTC-2MeSC-1 and CPTC-2MeSC, were developed and shown to have high immunoreactivity to unconjugated itaconate, itaconate-BSA conjugates, and Michael adducts of dimethyl itaconate. We found that CPTC-2MeSC-1 and CPTC-2MeSC-2 are specific and do not bind to other structurally similar cysteine Michael adducts, including those obtained from "sister" metabolites (fumarate, cis-aconitate), itaconate isomers (citraconate), and some itaconate esters. CPTC-2MeSC-2 is a useful tool in both studying biological actions of itaconate and developing therapeutic applications of itaconate and its derivatives.
Collapse
Affiliation(s)
- Lauren Switala
- Department
of Medicine, School of Medicine, Case Western
Reserve University, Cardiovascular Research Institute, Cleveland 44106-7078, United States
- Department
of Biomedical Engineering, Case Western
Reserve University, Cleveland 44106, United States
| | - Lin Di
- Department
of Medicine, School of Medicine, Case Western
Reserve University, Cardiovascular Research Institute, Cleveland 44106-7078, United States
- Department
of Biomedical Engineering, Case Western
Reserve University, Cleveland 44106, United States
| | - Simona Colantonio
- Cancer
Research Technology Program, Antibody Characterization Lab, Frederick National Laboratory for Cancer Research, Frederick, Maryland 21701, United States
| | - Bindu Lakshman
- Cancer
Research Technology Program, Antibody Characterization Lab, Frederick National Laboratory for Cancer Research, Frederick, Maryland 21701, United States
| | - Tessa W. Caceres
- Cancer
Research Technology Program, Antibody Characterization Lab, Frederick National Laboratory for Cancer Research, Frederick, Maryland 21701, United States
| | - Joshua J. Reading
- Cancer
Research Technology Program, Antibody Characterization Lab, Frederick National Laboratory for Cancer Research, Frederick, Maryland 21701, United States
| | - Sandra S. Garcia-Buntley
- Cancer
Research Technology Program, Antibody Characterization Lab, Frederick National Laboratory for Cancer Research, Frederick, Maryland 21701, United States
| | - Andrei Maiseyeu
- Department
of Medicine, School of Medicine, Case Western
Reserve University, Cardiovascular Research Institute, Cleveland 44106-7078, United States
- Department
of Biomedical Engineering, Case Western
Reserve University, Cleveland 44106, United States
| |
Collapse
|
50
|
He C, Chen P, Ning L, Huang X, Sun H, Wang Y, Zhao Y, Zeng C, Huang D, Gao H, Cao M. Inhibition of Mitochondrial Succinate Dehydrogenase with Dimethyl Malonate Promotes M2 Macrophage Polarization by Enhancing STAT6 Activation. Inflammation 2025:10.1007/s10753-024-02207-y. [PMID: 39806091 DOI: 10.1007/s10753-024-02207-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 11/13/2024] [Accepted: 12/03/2024] [Indexed: 01/16/2025]
Abstract
Macrophages exhibit diverse phenotypes depending on environment status, which contribute to physiological and pathological processes of immunological diseases, including sepsis, asthma, multiple sclerosis and colitis. The alternative activation of macrophages is tightly regulated to avoid excessive activation and damage of tissues and organs. Certain works characterized that succinate dehydrogenase (SDH) altered function of macrophages and promoted inflammatory response in M1 macrophages via mitochondrial reactive oxygen species (ROS). However, the effect of succinate dehydrogenase on M2 macrophage polarization remains incompletely understood. We employed dimethyl malonate (DMM) to inhibit succinate dehydrogenase activity and took use of RNA-seq to analyze the changes of inflammatory response of LPS-activated M1 macrophages or IL 4-activated M2 macrophages. Our data revealed that inhibition of SDH with DMM increased expression of M2 macrophages-associated signature genes, including Arg1, Ym1 and Mrc1. Consistent with previous work, we also observed that inhibition of SDH decreased the expression of IL-1β and enhanced the levels of IL-10 in M1 macrophages. Additionally, inhibition of SDH with DMM inhibited the production of chemokines, such as Cxcl3, Cxcl12, Ccl20 and Ccl9. DMM also amplified the M2 macrophages-related signature genes in IL-13-activated M2 macrophages. Mechanistic studies revealed that DMM promoted M2 macrophages polarization through mitochondrial ROS dependent STAT6 activation. Blocking ROS with mitoTEMPO or inhibiting STAT6 activation with ruxolitinib abrogated the promotion effect of DMM on M2 macrophages. Finally, dimethyl malonate treatment promoted peritoneal M2 macrophages differentiation and exacerbated OVA-induced allergy asthma in vivo. Collectively, we identified SDH as a braker to suppress M2 macrophage polarization via mitochondrial ROS, suggesting a novel strategy to treatment of M2 macrophages-mediated inflammatory diseases.
Collapse
Affiliation(s)
- Chaowen He
- Department of Respiratory Medicine, Shenzhen Longhua District Central Hospital, Shenzhen, 518110, China
| | - Pengfei Chen
- Department of Respiratory Medicine, Shenzhen Longhua District Central Hospital, Shenzhen, 518110, China
| | - Luwen Ning
- Health Science Center, Biobank, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen University, Shenzhen, China
| | - Xiuping Huang
- Department of Respiratory Medicine, Shenzhen Longhua District Central Hospital, Shenzhen, 518110, China
| | - Huimin Sun
- Department of Respiratory Medicine, Shenzhen Longhua District Central Hospital, Shenzhen, 518110, China
| | - Yuanyuan Wang
- Department of Respiratory Medicine, Shenzhen Longhua District Central Hospital, Shenzhen, 518110, China
| | - Yanli Zhao
- Department of Respiratory Medicine, Shenzhen Longhua District Central Hospital, Shenzhen, 518110, China
| | - Changchun Zeng
- Department of Respiratory Medicine, Shenzhen Longhua District Central Hospital, Shenzhen, 518110, China
| | - Dongsheng Huang
- Department of Respiratory Medicine, Shenzhen Longhua District Central Hospital, Shenzhen, 518110, China.
| | - Hanchao Gao
- Department of Nephrology, Shenzhen Longhua District Key Laboratory for Diagnosis and Treatment of Chronic Kidney Disease, Shenzhen Longhua District Central Hospital, Shenzhen, 518110, China.
| | - Mengtao Cao
- Department of Respiratory Medicine, Shenzhen Longhua District Central Hospital, Shenzhen, 518110, China.
| |
Collapse
|