1
|
Zhou X, Wang P, Xie L, Chan YK, Jiao Z, Shu R, Bai D, Lai S, Deng Y. Molybdoenzymes-emulating bio-heterojunction hydrogel with rapid disinfection and macrophage reprogramming for wound regeneration. Biomaterials 2025; 320:123284. [PMID: 40121831 DOI: 10.1016/j.biomaterials.2025.123284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 03/02/2025] [Accepted: 03/20/2025] [Indexed: 03/25/2025]
Abstract
Developing hydrogel dressings with the capabilities to accommodate irregular wounds and provide a cascade disinfective-regenerative microenvironment for wound repair is of great importance to combating pathogenic bacteria-infected wounds but remains an ongoing challenge. To address the conundrum, we devise a molybdoenzymes-emulating bio-heterojunction (M-bioHJ) doped double network (DN) hydrogel dressing for bacterial-infected wound healing. The near-infrared (NIR) photothermal effect of the M-bioHJ facilitates the exchange of multiple dynamic crosslinking sites in the hydrogel, endowing the hydrogel with photo-remote reprocessing capabilities to completely accommodate the encountered irregular wounds and ultimately accomplish the admirable therapeutic effect. Meanwhile, the introduced M-bioHJ shows NIR light-enhanced photodynamic activity to induce a massive engendering of reactive oxygen species (ROS), allowing rapid sterilization without reliance on exogenous hydrogen peroxide. Furthermore, the Mo ions released from the M-bioHJ-encapsulated hydrogel can play a crucial role in reprogramming the macrophage phenotype and determining tissue regeneration. Both in vitro and in vivo evidences authenticate the accelerated healing potential of infected wounds through the synergistic effects of photo-reprocessing, disinfection, and macrophage-reprogramming facilitated by the hydrogel. These findings highlight the promising application prospects of such neoteric M-bioHJ-encapsulated hydrogel dressings for wound disinfection and tissue regeneration.
Collapse
Affiliation(s)
- Xiong Zhou
- School of Chemical Engineering, West China Hospital of Stomatology, Sichuan University, Chengdu 610065, China; Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China
| | - Peiqi Wang
- School of Chemical Engineering, West China Hospital of Stomatology, Sichuan University, Chengdu 610065, China; State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics and Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Lu Xie
- School of Chemical Engineering, West China Hospital of Stomatology, Sichuan University, Chengdu 610065, China; State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics and Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Yau Kei Chan
- Department of Ophthalmology, The University of Hong Kong, 999077, Hong Kong, China
| | - Zheng Jiao
- Swanson School of Engineering, University of Pittsburgh, Pittsburgh, 15261, USA
| | - Rui Shu
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics and Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Ding Bai
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics and Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Shuangquan Lai
- School of Chemical Engineering, West China Hospital of Stomatology, Sichuan University, Chengdu 610065, China; Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Zhuhai, Guangdong 519000, China
| | - Yi Deng
- School of Chemical Engineering, West China Hospital of Stomatology, Sichuan University, Chengdu 610065, China; State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China; Department of Mechanical Engineering, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
2
|
Ioannidis M, Tjepkema J, Uitbeijerse MRP, van den Bogaart G. Immunomodulatory effects of 4-hydroxynonenal. Redox Biol 2025; 85:103719. [PMID: 40489926 DOI: 10.1016/j.redox.2025.103719] [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: 05/26/2025] [Accepted: 06/05/2025] [Indexed: 06/11/2025] Open
Abstract
The reactive aldehyde 4-hydroxy-2-nonenal (4-HNE) is a byproduct of lipid peroxidation driven by reactive oxygen species (ROS). 4-HNE covalently binds to macromolecules such as proteins, altering their functions. While 4-HNE is implicated in various ROS-related pathologies, its impact on the immune system remains incompletely understood. This review explores how 4-HNE influences molecular mechanisms involved in inflammation and immune cell functions. 4-HNE modulates inflammation through the interaction with several signaling pathways, including nuclear factor kappa-light-chain enhancement of activated B cells (NF-κB), nuclear factor erythroid 2-related factor (Nrf2), mitogen-activated protein kinases (MAPK), toll-like receptor (TLR) 4, and stimulator of interferon genes (STING), thereby affecting immune responses and modulating cytokine production and inflammasome activation. However, its effects are complex, exhibiting both pro- and anti-inflammatory properties depending on dose and cell type. This review highlights the multiple mechanisms by which 4-HNE modulates the immune cells' responses.
Collapse
Affiliation(s)
- Melina Ioannidis
- Department of Molecular Immunology, Groningen Biomolecular Science and Biotechnology Institute, University of Groningen, Nijenborgh 7, 9747AG, Groningen, the Netherlands
| | - Johanna Tjepkema
- Department of Molecular Immunology, Groningen Biomolecular Science and Biotechnology Institute, University of Groningen, Nijenborgh 7, 9747AG, Groningen, the Netherlands
| | - Michael R P Uitbeijerse
- Department of Molecular Immunology, Groningen Biomolecular Science and Biotechnology Institute, University of Groningen, Nijenborgh 7, 9747AG, Groningen, the Netherlands
| | - Geert van den Bogaart
- Department of Molecular Immunology, Groningen Biomolecular Science and Biotechnology Institute, University of Groningen, Nijenborgh 7, 9747AG, Groningen, the Netherlands; Department of Medical Biology and Pathology, University Medical Centre Groningen, Groningen, the Netherlands.
| |
Collapse
|
3
|
Zhang W, Meng L, Zhang X, Li Z, Hong F. CNPY2 drives DSS-induced colitis via the macrophage-ROS axis. Biomed Pharmacother 2025; 187:118078. [PMID: 40262233 DOI: 10.1016/j.biopha.2025.118078] [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: 02/12/2025] [Revised: 04/06/2025] [Accepted: 04/17/2025] [Indexed: 04/24/2025] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic inflammatory condition of the gastrointestinal tract, driven by genetic, environmental, and immune system factors. However, its exact mechanisms remain unclear. Here, we demonstrate that CNPY2 plays a critical role in colitis by modulating macrophage activity. Mice with whole-body Cnpy2 knockout (KO) exhibited significantly reduced dextran sodium sulfate (DSS)-induced colitis compared to wild-type controls. Cnpy2 KO mice showed less mucosal barrier disruption and fewer lamina propria macrophages (LPMφs) following DSS treatment. Proinflammatory cytokine production was also diminished in the colons of Cnpy2 KO mice. Furthermore, Cnpy2 KO macrophages generated markedly lower levels of reactive oxygen species (ROS), partly through CHOP regulation. Notably, treatment with the ROS scavenger N-acetyl-L-cysteine (NAC) completely abolished DSS-induced colitis in Cnpy2 KO mice. Thus, CNPY2 exacerbates DSS-induced colitis primarily through macrophage-specific effects, with ROS upregulation being central to its pathogenic role.
Collapse
Affiliation(s)
- Wenqing Zhang
- Pelotonia Institute for Immune-Oncology, The Ohio State University Comprehensive Cancer Center, United States; Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, 410 W 12th Ave, Columbus, OH 43210, United States
| | - Lingbin Meng
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, 410 W 12th Ave, Columbus, OH 43210, United States
| | - Xiaoli Zhang
- Biostatistics Core, College of Nursing, College of Public Health, University of South Florida, 4202 E Fowler Ave, Tampa, FL 33620, United States
| | - Zihai Li
- Pelotonia Institute for Immune-Oncology, The Ohio State University Comprehensive Cancer Center, United States; Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, 410 W 12th Ave, Columbus, OH 43210, United States.
| | - Feng Hong
- Pelotonia Institute for Immune-Oncology, The Ohio State University Comprehensive Cancer Center, United States; Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, 410 W 12th Ave, Columbus, OH 43210, United States.
| |
Collapse
|
4
|
He D, Dai W, Cui Y, Gao R, Yang X, Li W, Zhou J, Yin S, Kuang T, Zhu J, Luo L, Wang R, Qu Y, Yun C, Liu Z, Diao X, Ma X, Liang H, Wang F. Diamine oxidase acts as a novel risk factor in abnormal inflammation via mediating "cytosolic ROS-autophagy-IFN-γ" axis in NK cells. Life Sci 2025; 377:123775. [PMID: 40449879 DOI: 10.1016/j.lfs.2025.123775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Revised: 05/21/2025] [Accepted: 05/27/2025] [Indexed: 06/03/2025]
Abstract
AIMS Diamine oxidase (DAO), a well-established biomarker for intestinal damage, histamine intolerance or tumorigenesis, has rarely been reported in immune regulation. This study aimed to identify DAO as a critical enhancer of abnormal inflammation by promoting interferon-gamma (IFN-γ) production from natural killer (NK) cells. MAIN METHODS Clinical bioinformatics analyzed aoc1 (DAO-coding gene) expression in PBMCs from patients with inflammatory diseases. Murine models (LPS-induced systemic inflammation, sepsis, DSS-induced colitis) using DAO-/- mice, alongside DAO-/- NK92 cells and DAO inhibitor DIZE, were employed for phenotypic validation. Cellular profiling, bone marrow chimeras, reciprocal transplantation, RNA-sequence, non-targeted metabolomics, and flow cytometry were utilized to dissect DAO's mechanisms in NK cells. KEY FINDINGS DAO deficiency protected mice from inflammatory pathology by suppressing IFN-γ production. NK cells were identified as the primary target cells during the process, with DAO acting intracellularly to promote IFN-γ via a reactive oxygen species (ROS)-autophagy axis. DAO-derived ROS, distinct from mitochondrial or NOX2 sources, enhanced autophagic flux during NK activation, enabling IFN-γ biosynthesis. DAO did not affect NK homeostasis, including maturation, proliferation, or receptor expressions. SIGNIFICANCE DAO is a novel risk factor in inflammatory diseases, driving IFN-γ production through ROS-autophagy signaling in NK cells. Targeting DAO may offer therapeutic strategies for conditions involving dysregulated IFN-γ responses, including sepsis, colitis, and autoimmune disorders.
Collapse
Affiliation(s)
- Dongmei He
- The First Research Department, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center of PLA, Chongqing 400042, China
| | - Weihong Dai
- The First Research Department, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center of PLA, Chongqing 400042, China; Emergency of The Second Affiliated Hospital, Hainan Medical University, Haikou 571100, Hainan, China
| | - Yiqin Cui
- The First Research Department, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center of PLA, Chongqing 400042, China
| | - Rui Gao
- The First Research Department, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center of PLA, Chongqing 400042, China
| | - Xue Yang
- The First Research Department, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center of PLA, Chongqing 400042, China
| | - Wei Li
- The First Research Department, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center of PLA, Chongqing 400042, China
| | - Jing Zhou
- The First Research Department, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center of PLA, Chongqing 400042, China
| | - Shuangqin Yin
- The First Research Department, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center of PLA, Chongqing 400042, China
| | - Tianyin Kuang
- The First Research Department, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center of PLA, Chongqing 400042, China
| | - Junyu Zhu
- The First Research Department, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center of PLA, Chongqing 400042, China
| | - Li Luo
- The First Research Department, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center of PLA, Chongqing 400042, China
| | - Rixing Wang
- Emergency of The Second Affiliated Hospital, Hainan Medical University, Haikou 571100, Hainan, China
| | - Ye Qu
- Emergency of The Second Affiliated Hospital, Hainan Medical University, Haikou 571100, Hainan, China
| | - Caihong Yun
- Emergency of The Second Affiliated Hospital, Hainan Medical University, Haikou 571100, Hainan, China
| | - Zhuli Liu
- Emergency of The Second Affiliated Hospital, Hainan Medical University, Haikou 571100, Hainan, China
| | - Xiaoyan Diao
- The First Research Department, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center of PLA, Chongqing 400042, China
| | - Xiaoyuan Ma
- The First Research Department, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center of PLA, Chongqing 400042, China.
| | - Huaping Liang
- The First Research Department, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center of PLA, Chongqing 400042, China.
| | - Fangjie Wang
- The First Research Department, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center of PLA, Chongqing 400042, China.
| |
Collapse
|
5
|
Zhao C, Liu J, Zhao J, Wang C, Bai H, Yang Q, Ben J, Zhu X, Li X, Jiang B, Li K, Sun R, Ma X, Wang L, Zhang H, Chen Q. Monocyte to HDL cholesterol ratio predicts obesity-associated cardiac dysfunction. J Biomed Res 2025; 39:1-12. [PMID: 40432213 DOI: 10.7555/jbr.38.20240432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2025] Open
Abstract
As prevalence of obesity increases dramatically, obesity-associated cardiac dysfunction constitutes a huge challenge to human health. This study aims to find more useful lipid/inflammatory markers to predict the risk of obesity-associated cardiac dysfunction. By retrospectively analyzing the clinical characteristics of 5,648 disease patients, we find that both plasma level of high-density lipoprotein (HDL-C) and blood monocyte count have significant associations with left ventricular ejection fraction (LVEF) impairment. Univariate and multivariate regression analyses reveal that monocyte to HDL-C ratio (MHR) is stronger in predicting risk of LVEF decline than both HDL-C and monocyte. Mediation analysis further indicates a mediative effect of high MHR on the obesity-associated cardiac systolic function decline. As such, our results demonstrate a superior role of MHR in predicting risk of obesity-associated decline in cardiac systolic function among the routine metabolic/inflammatory markers.
Collapse
Affiliation(s)
- Chunsheng Zhao
- Department of Pathophysiology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Jinting Liu
- Department of Pathophysiology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Key Laboratory of Jiangsu Province on Targeted Intervention of Cardiovascular Diseases, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Jiaqi Zhao
- Department of Pathophysiology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Key Laboratory of Jiangsu Province on Targeted Intervention of Cardiovascular Diseases, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Chao Wang
- Key Lab of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Hui Bai
- Department of Pathophysiology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Key Laboratory of Jiangsu Province on Targeted Intervention of Cardiovascular Diseases, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Qing Yang
- Department of Pathophysiology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Key Laboratory of Jiangsu Province on Targeted Intervention of Cardiovascular Diseases, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Jingjing Ben
- Department of Pathophysiology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Key Laboratory of Jiangsu Province on Targeted Intervention of Cardiovascular Diseases, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Xudong Zhu
- Department of Pathophysiology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Key Laboratory of Jiangsu Province on Targeted Intervention of Cardiovascular Diseases, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Xiaoyu Li
- Department of Pathophysiology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Key Laboratory of Jiangsu Province on Targeted Intervention of Cardiovascular Diseases, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Bin Jiang
- Department of Pathophysiology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Key Laboratory of Jiangsu Province on Targeted Intervention of Cardiovascular Diseases, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Kai Li
- Donghai County People's Hospital, Donghai, Jiangsu 222300, China
| | - Runfeng Sun
- Donghai County People's Hospital, Donghai, Jiangsu 222300, China
| | - Xuexing Ma
- Department of Cardiology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu 215006, China
| | - Liansheng Wang
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Hanwen Zhang
- Department of Pathophysiology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Key Laboratory of Jiangsu Province on Targeted Intervention of Cardiovascular Diseases, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Qi Chen
- Department of Pathophysiology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Key Laboratory of Jiangsu Province on Targeted Intervention of Cardiovascular Diseases, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| |
Collapse
|
6
|
Chen D, Guo Z, Yao L, Sun Y, Dian Y, Zhao D, Ke Y, Zeng F, Zhang C, Deng G, Li L. Targeting oxidative stress-mediated regulated cell death as a vulnerability in cancer. Redox Biol 2025; 84:103686. [PMID: 40424719 DOI: 10.1016/j.redox.2025.103686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2025] [Accepted: 05/17/2025] [Indexed: 05/29/2025] Open
Abstract
Reactive oxygen species (ROS), regulators of cellular behaviors ranging from signaling to cell death, have complex production and control mechanisms to maintain a dynamic redox balance under physiological conditions. Redox imbalance is frequently observed in tumor cells, where ROS within tolerable limits promote oncogenic transformation, while excessive ROS induce a range of regulated cell death (RCD). As such, targeting ROS-mediated regulated cell death as a vulnerability in cancer. However, the precise regulatory networks governing ROS-mediated cancer cell death and their therapeutic applications remain inadequately characterized. In this Review, we first provide a comprehensive overview of the mechanisms underlying ROS production and control within cells, highlighting their dynamic balance. Next, we discuss the paradoxical nature of the redox system in tumor cells, where ROS can promote tumor growth or suppress it, depending on the context. We also systematically explored the role of ROS in tumor signaling pathways and revealed the complex ROS-mediated cross-linking networks in cancer cells. Following this, we focus on the intricate regulation of ROS in RCD and its current applications in cancer therapy. We further summarize the potential of ROS-induced RCD-based therapies, particularly those mediated by drugs targeting specific redox balance mechanisms. Finally, we address the measurement of ROS and oxidative damage in research, discussing existing challenges and future prospects of targeting ROS-mediated RCD in cancer therapy. We hope this review will offer promise for the clinical application of targeting oxidative stress-mediated regulated cell death in cancer therapy.
Collapse
Affiliation(s)
- Danyao Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, China; Furong Laboratory, Changsha, Hunan, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, China; Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ziyu Guo
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, China; Furong Laboratory, Changsha, Hunan, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, China
| | - Lei Yao
- Department of Liver Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yuming Sun
- Department of Plastic and Cosmetic Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan Province, China
| | - Yating Dian
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, China; Furong Laboratory, Changsha, Hunan, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, China
| | - Deze Zhao
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yizhe Ke
- The First Affliated Hospital of Shihezi University, China
| | - Furong Zeng
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Chunfang Zhang
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Guangtong Deng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, China; Furong Laboratory, Changsha, Hunan, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, China.
| | - Linfeng Li
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
7
|
Zhou H, Lao M, Liang Z, Zhao H, Wang Y, Huang Q, Ou C. Identification of M0 macrophage associated lipid metabolism genes for prognostic and immunotherapeutic response prediction in hepatocellular carcinoma. Discov Oncol 2025; 16:781. [PMID: 40377731 DOI: 10.1007/s12672-025-02620-1] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 05/08/2025] [Indexed: 05/18/2025] Open
Abstract
PURPOSE Liver cancer prognosis is associated with M0 macrophages and lipid metabolism reprogramming; however, the prognostic role of M0 macrophage-related lipid metabolism genes in hepatocellular carcinoma (HCC) remains unclear. METHODS We identified 153 lipid metabolism genes associated with M0 macrophage infiltration in HCC from The Cancer Genome Atlas (TCGA) and the Molecular Signatures Database (MSigDB). Prognostic genes were selected, and a model was constructed using least absolute shrinkage and selection operator (LASSO) and Cox regression analyses. The model was validated using the International Cancer Genome Consortium (ICGC) database. We assessed the expression levels of prognostic genes by quantitative real-time polymerase chain reaction (qRT‒PCR). RESULTS A prognostic model was developed based on five characteristic genes. Receiver operating characteristic curve analysis demonstrated that the model had good accuracy, with area under the curve values of 0.796, 0.732, and 0.728 for predicting survival at 1, 3, and 5 years, respectively. The high-risk group exhibited increased sensitivity to common chemotherapy drugs, including sorafenib, dasatinib, and 5-fluorouracil, compared with the low-risk group (P < 0.05). Additionally, the high-risk group had significantly more infiltrating M0 macrophages, resting dendritic cells, follicular helper T cells, and regulatory T cells than did the low-risk group (P < 0.05). The qRT‒PCR results confirmed the upregulation of these five characteristic genes in HCC tissues. CONCLUSIONS M0 macrophage-associated lipid metabolism genes may serve as biomarkers for the prognosis of patients with HCC and as targets for immunotherapy.
Collapse
Affiliation(s)
- Huanjie Zhou
- Department of Clinical Laboratory, Guangxi Medical University Cancer Hospital, Guangxi Zhuang Autonomous Region, Nanning, 530021, People's Republic of China
| | - Ming Lao
- Department of Clinical Laboratory, Guangxi Medical University Cancer Hospital, Guangxi Zhuang Autonomous Region, Nanning, 530021, People's Republic of China
| | - Zhengui Liang
- Department of Clinical Laboratory, Guangxi Medical University Cancer Hospital, Guangxi Zhuang Autonomous Region, Nanning, 530021, People's Republic of China
| | - Huiliu Zhao
- Department of Clinical Laboratory, Guangxi Medical University Cancer Hospital, Guangxi Zhuang Autonomous Region, Nanning, 530021, People's Republic of China
| | - Ying Wang
- Department of Clinical Laboratory, Guangxi Medical University Cancer Hospital, Guangxi Zhuang Autonomous Region, Nanning, 530021, People's Republic of China
| | - Qiongqing Huang
- Department of Clinical Laboratory, Guangxi Medical University Cancer Hospital, Guangxi Zhuang Autonomous Region, Nanning, 530021, People's Republic of China
| | - Chao Ou
- Department of Clinical Laboratory, Guangxi Medical University Cancer Hospital, Guangxi Zhuang Autonomous Region, Nanning, 530021, People's Republic of China.
| |
Collapse
|
8
|
Wei J, Fu Y, Feng S, Zhang J, Zhang Y, Yu J, Kang P, Wu C, Mi H. The Effects of Fishmeal Replacement with Degossypolled Cottonseed Protein on Growth, Serum Biochemistry, Endocrine Responses, Lipid Metabolism, and Antioxidant and Immune Responses in Black Carp ( Mylopharyngodon piceus). Animals (Basel) 2025; 15:1404. [PMID: 40427281 PMCID: PMC12108466 DOI: 10.3390/ani15101404] [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: 03/03/2025] [Revised: 04/14/2025] [Accepted: 04/23/2025] [Indexed: 05/29/2025] Open
Abstract
This research investigated the growth, serum biochemistry, antioxidant capability, and immunity impact of black carp fed degossypolled cottonseed protein replacing fishmeal at the levels of 0%, 10%, 20%, 30%, 40%, and 50% (DCP0, DCP10, DCP20, DCP30, DCP40, and DCP50), respectively. The results showed there were no significant changes in growth among these test groups. The activities and mRNA expression levels of amylase and trypsin were heightened in conjunction with 30-40% DCP. Although the insulin contents were reduced with a rise in DCP content, 5-hydroxytryptamino was increased in the DCP40 and DCP50 groups. DCP40 could heighten the levels of low-density lipoprotein cholesterol, triglycerides, total cholesterol, and urea nitrogen. Although lower levels of DCP (≤20%) could increase the total antioxidant capacity compared with the DCP50 group, DCP50 could markedly heighten levels of catalase, glutathione S-transferase, H2O2, and malondialdehyde. Meanwhile, the mRNA levels of Mn-superoxide dismutase, glutathione reductase, glutathione peroxidase, glutamate-cysteine ligase regulatory subunit, and nuclear factor E2-related factor 2 were heightened in the DCP30 group compared with the DCP50 group. The levels of alkaline phosphatase, immunoglobulin M, and liver-expressed antimicrobial peptide 2 were markedly heightened in the liver of the DCP20 group compared with the DCP50 group. In conclusion, a suitable level of DCP (20%) could improve serum biochemical indices and hormone variation, enhance antioxidant capability, and increase immunity in black carp.
Collapse
Affiliation(s)
- Jiao Wei
- National-Local Joint Engineering Laboratory of Aquatic Animal Genetic Breeding and Nutrition (Zhejiang), Huzhou University, 759 East 2nd Road, Huzhou 313000, China; (J.W.); (Y.F.); (S.F.); (J.Z.); (Y.Z.); (J.Y.)
| | - Yifan Fu
- National-Local Joint Engineering Laboratory of Aquatic Animal Genetic Breeding and Nutrition (Zhejiang), Huzhou University, 759 East 2nd Road, Huzhou 313000, China; (J.W.); (Y.F.); (S.F.); (J.Z.); (Y.Z.); (J.Y.)
| | - Shinan Feng
- National-Local Joint Engineering Laboratory of Aquatic Animal Genetic Breeding and Nutrition (Zhejiang), Huzhou University, 759 East 2nd Road, Huzhou 313000, China; (J.W.); (Y.F.); (S.F.); (J.Z.); (Y.Z.); (J.Y.)
| | - Jinjing Zhang
- National-Local Joint Engineering Laboratory of Aquatic Animal Genetic Breeding and Nutrition (Zhejiang), Huzhou University, 759 East 2nd Road, Huzhou 313000, China; (J.W.); (Y.F.); (S.F.); (J.Z.); (Y.Z.); (J.Y.)
| | - Yuanyuan Zhang
- National-Local Joint Engineering Laboratory of Aquatic Animal Genetic Breeding and Nutrition (Zhejiang), Huzhou University, 759 East 2nd Road, Huzhou 313000, China; (J.W.); (Y.F.); (S.F.); (J.Z.); (Y.Z.); (J.Y.)
| | - Jiaxing Yu
- National-Local Joint Engineering Laboratory of Aquatic Animal Genetic Breeding and Nutrition (Zhejiang), Huzhou University, 759 East 2nd Road, Huzhou 313000, China; (J.W.); (Y.F.); (S.F.); (J.Z.); (Y.Z.); (J.Y.)
| | - Pengtian Kang
- Gansu Provincial Aquatic Technology Extension Station, 113 Zhongshan Road, Lanzhou 730030, China
| | - Chenglong Wu
- National-Local Joint Engineering Laboratory of Aquatic Animal Genetic Breeding and Nutrition (Zhejiang), Huzhou University, 759 East 2nd Road, Huzhou 313000, China; (J.W.); (Y.F.); (S.F.); (J.Z.); (Y.Z.); (J.Y.)
| | - Haifeng Mi
- Healthy Aquaculture Key Laboratory of Sichuan Province, Tongwei Co., Ltd., 588 Tianfu Avenue, Chengdu 610093, China;
| |
Collapse
|
9
|
Song Y, Li H, Yuan Y, Zhang D, Wang Z, Qi B, Jiang P, Yu A. Synergistic photothermal-sonodynamic therapy for antibacterial and immune reprogramming in chronic osteomyelitis. J Control Release 2025; 381:113612. [PMID: 40073945 DOI: 10.1016/j.jconrel.2025.113612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 03/04/2025] [Accepted: 03/06/2025] [Indexed: 03/14/2025]
Abstract
The development of antibiotic resistance and inadequate immune response in chronic inflammation pose significant challenges in treating chronic osteomyelitis. As accepted non-antibiotic antimicrobial therapies, sonodynamic therapy (SDT) and photothermal therapy (PTT) are recognized for their effectiveness in eliminating bacteria and promoting tissue repair, rendering them promising therapeutic strategies for treating bacterial infections and preventing the emergence of drug-resistant bacteria. However, the antimicrobial action and efficacy in promoting tissue repair depend on the activation status of the host immune system. In this study, by encapsulating horseradish peroxidase (HRP)-loaded gold/polydopamine (PDA) nanoparticles within DOTAP/DOPE cationic liposomes (DLPs), a novel multifunctional nanocatalyst, Au/PDA/HRP@DLP (APH@DLP), was developed to achieve antimicrobial effects and immunological reprogramming of chronic osteomyelitis through synergistic SDT and PTT. The impact on immune activation was investigated by assessing the anti-infective and healing effects in osteomyelitis rat models. The release of bacterial-associated antigens during treatment serves as an in situ vaccine, activating antigen-presenting cells and further stimulating adaptive immunity, while also inducing immune memory that significantly reduces the risk of recurrence. Additionally, macrophage phenotypic transformation during SDT and PTT facilitates tissue repair. This study highlights the role of immune activation in SDT/PTT-based antimicrobial therapy and suggests new strategies for treating chronic osteomyelitis.
Collapse
Affiliation(s)
- Yuchen Song
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Haimei Li
- School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China; Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (MOE), Wuhan University, Wuhan 430072, China
| | - Ying Yuan
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Dong Zhang
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Zheng Wang
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Baiwen Qi
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China.
| | - Peng Jiang
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China; Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (MOE), Wuhan University, Wuhan 430072, China.
| | - Aixi Yu
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China.
| |
Collapse
|
10
|
Zhao Y, Wang Y, Liang T, Song X, Zhu Y, Liu X, Lv M, Zheng C, Ni F. Dysregulated glutathione metabolism impairs natural killer cell function in patients with acute leukemia. Int Immunopharmacol 2025; 154:114566. [PMID: 40184815 DOI: 10.1016/j.intimp.2025.114566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 03/11/2025] [Accepted: 03/25/2025] [Indexed: 04/07/2025]
Abstract
Natural killer (NK) cell function is markedly impaired in patients with acute leukemia, weakening their anti-tumor immune response. However, the mechanisms underlying NK cell dysfunction are not fully understood. Here, we reveal that NK cells from patients with acute leukemia (AL-NK) exhibit significantly reduced intracellular glutathione (GSH) levels, accompanied by disrupted redox homeostasis and increased levels of mitochondrial reactive oxygen species. Flow cytometry and transcriptomic analyses indicate that dysregulated GSH metabolism leads to mitochondrial dysfunction in NK cells, thereby impairing their antileukemic cytotoxicity and proliferative capacity. Notably, supplementation with glutathione reduced ethyl ester (GSHEE)-a GSH precursor-effectively restores GSH levels in AL-NK cells, enhancing mitochondrial activity, oxidative phosphorylation, ATP production, and NK cell-mediated cytotoxicity. Moreover, GSHEE treatment activates the mTOR signaling pathway in NK cells, further promoting their function and proliferation. Overall, our study identifies dysregulated GSH metabolism as a key driver of NK cell dysfunction in acute leukemia and suggests that GSH-based interventions may provide a promising strategy to enhance NK cell-mediated immunotherapies.
Collapse
Affiliation(s)
- Yue Zhao
- Department of Hematology, The First Affiliated Hospital of USTC, National Key Laboratory of Immune Response and Immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Institute of Blood and Cell Therapy and Anhui Provincial Key Laboratory of Blood Research and Applications, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; Institute of Immunology, The CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230022, China
| | - Yan Wang
- Department of Hematology, The First Affiliated Hospital of USTC, National Key Laboratory of Immune Response and Immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Institute of Blood and Cell Therapy and Anhui Provincial Key Laboratory of Blood Research and Applications, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; Department of Clinical Laboratory, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China
| | - Tingting Liang
- Department of Hematology, The First Affiliated Hospital of USTC, National Key Laboratory of Immune Response and Immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Institute of Blood and Cell Therapy and Anhui Provincial Key Laboratory of Blood Research and Applications, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; Institute of Immunology, The CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230022, China
| | - Xian Song
- Department of Hematology, The First Affiliated Hospital of USTC, National Key Laboratory of Immune Response and Immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Institute of Blood and Cell Therapy and Anhui Provincial Key Laboratory of Blood Research and Applications, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; Institute of Immunology, The CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230022, China
| | - Yingqiao Zhu
- Department of Hematology, The First Affiliated Hospital of USTC, National Key Laboratory of Immune Response and Immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Institute of Blood and Cell Therapy and Anhui Provincial Key Laboratory of Blood Research and Applications, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Xinru Liu
- Department of Hematology, The First Affiliated Hospital of USTC, National Key Laboratory of Immune Response and Immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Institute of Blood and Cell Therapy and Anhui Provincial Key Laboratory of Blood Research and Applications, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; Institute of Immunology, The CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230022, China
| | - Mengya Lv
- Department of Hematology, The First Affiliated Hospital of USTC, National Key Laboratory of Immune Response and Immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Institute of Blood and Cell Therapy and Anhui Provincial Key Laboratory of Blood Research and Applications, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Changcheng Zheng
- Department of Hematology, The First Affiliated Hospital of USTC, National Key Laboratory of Immune Response and Immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Institute of Blood and Cell Therapy and Anhui Provincial Key Laboratory of Blood Research and Applications, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China.
| | - Fang Ni
- Department of Hematology, The First Affiliated Hospital of USTC, National Key Laboratory of Immune Response and Immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Institute of Blood and Cell Therapy and Anhui Provincial Key Laboratory of Blood Research and Applications, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; Institute of Immunology, The CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230022, China.
| |
Collapse
|
11
|
Meenatchi V, Won SY, Almarhoon ZM, Han SS. Assessment of pyrazolone derivatives as a new class of cyclooxygenase-2 and aflatoxigenic fungal inhibitors. Int J Biol Macromol 2025; 311:143903. [PMID: 40334893 DOI: 10.1016/j.ijbiomac.2025.143903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Revised: 04/23/2025] [Accepted: 05/01/2025] [Indexed: 05/09/2025]
Abstract
The development of anti-inflammatory compounds is crucial because of the complexity of the wound-healing process associated with bacterial infections. Anti-inflammatory compounds have been rapidly developed for use in biomedical fields. In this study, three new skin fibroblast cell promotors (pyrazolone derivatives) with pyrazolone cores were designed and synthesized to evaluate their inhibitory effects on the cyclooxygenase-2 enzyme. The pyrazolone derivatives were characterized using FTIR, 1H and 13C NMR, and DSC. These pyrazolone derivatives exhibited excellent biocompatibility, resulting in significant proliferation of NIH/3 T3 fibroblast cells, as confirmed through fluorescence microscopy in the live-dead cell assay. Moral anti-inflammatory property was verified by the strong interactions between inflammation-responsible enzyme 6-COX and ligands. In vitro anticancer activity assessments revealed that the pyrazolone derivatives did not reduce the viability of breast cancer MDA-MB231 cells as opposed to the control group. Additionally, the pyrazolone derivatives displayed great antibacterial activity against Staphylococcus aureus and Escherichia coli, with a 100 ± 0.1 % inhibitory efficiency for 24 h. Moreover, the pyrazolone derivatives showed good antifungal activity against Aspergillus flavus, effectively inhibiting the secretion of aflatoxins (98 ± 0.1 %). Our results demonstrated that these newly synthesized pyrazolone derivatives are promising candidates for application in anti-inflammatory or wound-dressing treatments.
Collapse
Affiliation(s)
- Venkatasamy Meenatchi
- School of Chemical Engineering, Yeungnam University, Gyeongsan-si, Gyeongsangbuk-do 38541, Republic of Korea.
| | - So Yeon Won
- School of Chemical Engineering, Yeungnam University, Gyeongsan-si, Gyeongsangbuk-do 38541, Republic of Korea
| | - Zainab M Almarhoon
- Department of Chemistry, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Sung Soo Han
- School of Chemical Engineering, Yeungnam University, Gyeongsan-si, Gyeongsangbuk-do 38541, Republic of Korea.
| |
Collapse
|
12
|
Dong J, Li Z, Fu C, Yang D, Yang H, Lin L, Liang XJ, Chen Z, Chen L, Guo W. Cardiosplenic axis-targeted immunomodulatory liposome for myocardial ischemia-reperfusion injury treatment. J Control Release 2025; 383:113799. [PMID: 40324533 DOI: 10.1016/j.jconrel.2025.113799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 04/02/2025] [Accepted: 04/28/2025] [Indexed: 05/07/2025]
Abstract
Monocyte/macrophage (Mo/Mϕ) infiltration is critical in myocardial ischemia-reperfusion injury (MIRI). However, the complex composition of the myocardium severely hinders drug accumulation and makes it challenging to modulate the Mo/Mϕ immune response at the MIRI site. The spleen, acting as a Mo/Mϕ reservoir, plays a crucial role in the development of MIRI along the cardiosplenic axis. Compared to directly delivering medications to the MIRI site, targeting the spleen for Mo/Mϕ immunomodulation provides an alternative strategy to modulate the immunological phenotype on-site. Therefore, we developed a melatonin-loaded liposome (ST-MT@lipo2) that specifically targets the spleen and can effectively regulate the immunological response of splenic monocytes and macrophages, consequently enhancing their immune response at the site of MIRI. Additionally, the splenectomy mouse model revealed that ST-MT@lipo2 regulated MIRI's immune response through the cardiosplenic axis by regulating the MCP-1/CCR2 pathway to reduce circulating inflammatory monocyte migration from the spleen to the MIRI site. Moreover, pathological staining and echocardiography showed that ST-MT@lipo2 reduced myocardial damage and improved cardiac function in MIRI mice. This study demonstrates the crucial importance of modulating the immune response in the cardiosplenic axis for treating MIRI, which also inspired the treatments for inflammatory diseases by controlling the spleen immunological milieu.
Collapse
Affiliation(s)
- Jiankai Dong
- Department of Cardiology, Fujian Medical University Union Hospital, Fuzhou 250001, China
| | - Zechuan Li
- Department of Minimally Invasive Interventional Radiology, The Second Affiliated Hospital, School of Biomedical Engineering, Guangzhou Medical University, Guangzhou 510260, China
| | - Chenxing Fu
- Department of Minimally Invasive Interventional Radiology, The Second Affiliated Hospital, School of Biomedical Engineering, Guangzhou Medical University, Guangzhou 510260, China
| | - Ding Yang
- Department of Radiology, Peking University Cancer Hospital & Institute, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Beijing 100142, China
| | - Huijie Yang
- Department of Neurology, The Fifth Affiliated Hospital of Southern Medical University, Guangzhou 510260, China
| | - Li Lin
- Department of Cardiology, Fujian Medical University Union Hospital, Fuzhou 250001, China
| | - Xing-Jie Liang
- Department of Minimally Invasive Interventional Radiology, The Second Affiliated Hospital, School of Biomedical Engineering, Guangzhou Medical University, Guangzhou 510260, China; Laboratory of Controllable Nanopharmaceuticals, Chinese Academy of Sciences (CAS) Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing 100190, China
| | - Zhaoyang Chen
- Department of Cardiology, Fujian Medical University Union Hospital, Fuzhou 250001, China.
| | - Lianglong Chen
- Department of Cardiology, Fujian Medical University Union Hospital, Fuzhou 250001, China.
| | - Weisheng Guo
- Department of Minimally Invasive Interventional Radiology, The Second Affiliated Hospital, School of Biomedical Engineering, Guangzhou Medical University, Guangzhou 510260, China.
| |
Collapse
|
13
|
Francois AA, Yin X, Oka S, Sadoshima J, Mayr M, Eaton P. On the utility of immobilized phenylarsine oxide in the study of redox sensitive cardiac proteins. Sci Rep 2025; 15:15554. [PMID: 40319072 PMCID: PMC12049532 DOI: 10.1038/s41598-025-00665-4] [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: 01/24/2025] [Accepted: 04/29/2025] [Indexed: 05/07/2025] Open
Abstract
Reactive protein cysteine thiols are critical to sensing and transducing oxidant signals, often by induction of disulfide bonds that alter their activity or interactions. Identifying such redox active proteins nowadays is mostly achieved using thiol redox proteomics with such datasets increasingly available. Subsequently, we are challenged with determining how changes in the redox state of a protein of interest alters its activity or interactions and how this affects physiology or disease progression including in vivo scenarios. Such studies necessitate the measurement of how the protein redox state changes with health or disease-related interventions, with it not always being practicable to resort back to resource-intensive proteomics to achieve this. In some proteins, oxidation to a disulfide state causes a non-reducing gel-shift, but this is mostly not the case and so other efficient approaches are required to index changes in redox state. Here we assessed the utility of immobilized, solid-phase phenylarsine oxide (PAO-Sepharose) as a tool for indexing the thiol redox state of candidate proteins in cardiac samples from in vivo interventions associated with oxidative stress. PAO-Sepharose, which binds proteins with proximal reduced thiol pairs but not when they form a disulfide, was also used to identify proteins that that are oxidised in isolated perfused mouse hearts exposed to hydrogen peroxide or diamide using proteomics. This together with complementary studies using a cardiac-specific FLAG-Thioredoxin-1C35S-HA transgenic 'trap-mutant' mouse model allowed identification of heart proteins susceptible to oxidant-induced disulfide bond formation using proteomics. Thus, two in vitro approaches identified putative cardiac thiol redox sensor proteins that were then assessed with in vivo follow-up studies for their susceptibility to oxidation during endotoxemia induced by lipopolysaccharide or type I diabetes induced by streptozotocin in mice. Of five proteins selected for further analysis by PAO-Sepharose binding, two, namely apoptotic protease activating factor 1 interacting protein (APIP) and γ-glutamylcyclotransferase (GGCT), displayed significantly lower affinity capture from hearts from lipopolysaccharide- or streptozotocin-treated mice, consistent with oxidation of their vicinal thiols. We conclude that PAO-Sepharose is an effective and accessible tool for identifying oxidant-sensitive protein thiols in both ex vivo and in vivo models of oxidative stress. As increasing numbers of thiol redox proteins are identified, PAO-Sepharose binding is an efficient method to determine if they change their oxidation state during interventions relevant to health and disease.
Collapse
Affiliation(s)
- Asvi Arora Francois
- William Harvey Research Institute, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK.
| | - Xiaoke Yin
- National Heart and Lung Institute, Imperial College London, London, SW7 2AZ, UK
| | - Shinichi Oka
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, NJ, 07101, USA
| | - Junichi Sadoshima
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, NJ, 07101, USA
| | - Manuel Mayr
- National Heart and Lung Institute, Imperial College London, London, SW7 2AZ, UK
| | - Philip Eaton
- William Harvey Research Institute, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK.
| |
Collapse
|
14
|
Han S, Wan J, Zhang X, Ding J, Li X, Cheng Y, Sun Y, Xu Z, Wu J, Chen R. Proteomic profiling of spleen in rat infected with clonorchis sinensis using liquid chromatography tandem mass spectrometry analysis. Acta Trop 2025; 265:107594. [PMID: 40127806 DOI: 10.1016/j.actatropica.2025.107594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 03/09/2025] [Accepted: 03/20/2025] [Indexed: 03/26/2025]
Abstract
Clonorchiasis, caused by Clonorchis sinensis, remains a significant neglected tropical disease with substantial global health implications. As the largest immune organ in mammals, the spleen plays a crucial role in defending against C. sinensis infection; however, the molecular mechanisms underlying spleen pathogenesis during such infections remain poorly understood. To address this gap, quantitative Tandem Mass Tags (TMT) liquid chromatography-tandem mass spectrometry was employed to profile protein changes in the spleens of rats infected with C. sinensis. This analysis identified 40,664 peptides from 6817 proteins, including 371 and 464 differentially expressed proteins at 4 and 8 weeks post-infection (wpi) compared to the control groups, respectively. Clustering analysis revealed distinct proteomic profiles among the groups, while gene ontology analysis associated the differentially expressed proteins with biological binding activities and metabolic processes. KEGG analysis revealed significant enrichment of immune-related and metabolic pathways, including AMPK, IL-17, and p53 signaling pathways. These findings reveal dynamic proteomic alterations in the spleen during C. sinensis infection, offering valuable insights into the biomarker candidates for early diagnosis. Further studies are warranted to validate these potential biomarkers and explore their utility for early diagnosis of clonorchiasis.
Collapse
Affiliation(s)
- Su Han
- Department of Public Health and Preventive Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, China; Department of Parasitology, Harbin Medical University, Harbin, China; Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Jie Wan
- Department of Public Health and Preventive Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Xiaoli Zhang
- Department of Parasitology, Harbin Medical University, Harbin, China
| | - Jian Ding
- Department of Parasitology, Harbin Medical University, Harbin, China
| | - Xiang Li
- Department of Parasitology, Harbin Medical University, Harbin, China
| | - Yang Cheng
- Department of Public Health and Preventive Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Yifan Sun
- Department of Public Health and Preventive Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Zhenli Xu
- Department of Public Health and Preventive Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Jianlin Wu
- Wuming Hospital of Guangxi medical university, Nanning, China.
| | - Rui Chen
- Affiliated Hospital of Jiangnan University, Wuxi, China.
| |
Collapse
|
15
|
Banki K, Perl A. Cell type-specific regulation of the pentose phosphate pathway during development and metabolic stress-driven autoimmune diseases: Relevance for inflammatory liver, renal, endocrine, cardiovascular and neurobehavioral comorbidities, carcinogenesis, and aging. Autoimmun Rev 2025; 24:103781. [PMID: 40010622 DOI: 10.1016/j.autrev.2025.103781] [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: 02/11/2025] [Revised: 02/19/2025] [Accepted: 02/20/2025] [Indexed: 02/28/2025]
Abstract
The pathogenesis of autoimmunity is incompletely understood which limits the development of effective therapies. New compelling evidence indicates that the pentose phosphate pathway (PPP) profoundly regulate lineage development in the immune system that are influenced by genetic and environmental factors during metabolic stress underlying the development of autoimmunity. The PPP provides two unique metabolites, ribose 5-phosphate for nucleotide biosynthesis in support of cell proliferation and NADPH for protection against oxidative stress. The PPP operates two separate branches, oxidative (OxPPP) and non-oxidative (NOxPPP). While the OxPPP functions in all organisms, the NOxPPP reflects adaptation to niche-specific metabolic requirements. The OxPPP primarily depends on glucose 6-phosphate dehydrogenase (G6PD), whereas transaldolase (TAL) controls the rate and directionality of metabolic flux though the NOxPPP. G6PD is essential for normal development but its partial deficiency protects from malaria. Although men and mice lacking TAL develop normally, they exhibit liver cirrhosis progressing to hepatocellular carcinoma. Mechanistic target of rapamycin-dependent loss of paraoxonase 1 drives autoimmunity and cirrhosis in TAL deficiency, while hepatocarcinogenesis hinges on polyol pathway activation via aldose reductase (AR). Accumulated polyols, such as erythritol, xylitol, and sorbitol, which are commonly used as non-caloric sweeteners, may act as pro-inflammatory oncometabolites under metabolic stress, such as TAL deficiency. The TAL/AR axis is identified as a checkpoint of pathogenesis and target for treatment of metabolic stress-driven systemic autoimmunity with relevance for inflammatory liver, renal and cardiovascular disorders, diabetes, carcinogenesis, and aging.
Collapse
Affiliation(s)
- Katalin Banki
- Departments of Medicine, Microbiology and Immunology, Biochemistry and Molecular Biology, and Pathology, State University of New York Upstate Medical University, Norton College of Medicine, 750 East Adams Street, Syracuse, NY 13210, USA
| | - Andras Perl
- Departments of Medicine, Microbiology and Immunology, Biochemistry and Molecular Biology, and Pathology, State University of New York Upstate Medical University, Norton College of Medicine, 750 East Adams Street, Syracuse, NY 13210, USA.
| |
Collapse
|
16
|
Yan Y, Fang M, Zhao C, Lin X, Tong C, Xiang C, Ran Y, Wang X, Li S, Chen G, Fu L. Dl-3-n-butylphthalide attenuates DOX-induced cardiotoxicity in mice by inhibiting Nrf2/Keap1 complex formation. Front Pharmacol 2025; 16:1542296. [PMID: 40365306 PMCID: PMC12069325 DOI: 10.3389/fphar.2025.1542296] [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: 12/09/2024] [Accepted: 04/21/2025] [Indexed: 05/15/2025] Open
Abstract
Introduction Drug-induced cardiotoxicity (DICT), defined as myocardial injury caused by direct or indirect toxicity of therapeutic agents, disrupts cardiovascular homeostasis, underscoring the urgent need for preventive strategies in clinical practice. Doxorubicin (DOX), a clinically established anthracycline chemotherapeutic, induces dose-dependent cardiotoxicity driven by reactive oxygen species overproduction. Notably, Dl-3-n-butylphthalide (NBP), a bioactive phytochemical derived from celery, has shown potential in mitigating DOX-induced cardiomyopathy via its antioxidant activity. Therefore, this study aimed to investigate the protective effects of NBP on DOX-induced cardiomyopathy, with a focus on elucidating the underlying mechanisms. Method We developed both in vivo and in vitro models of DOX-induced cardiotoxicity. For the animal model, male C57BL/6 mice were administered with DOX (4 mg/kg, i.p.) once a week for 3 weeks. For the cell model, H9C2 myoblasts were exposed to 1 μM DOX for at least 6 h to establish acute cardiotoxicity. Results Our results demonstrate that NBP significantly improves cardiac function, as evidenced by approximately 10% increase in cardiac functional parameters (ejection fraction and left ventricular shortening fraction). Besides, NBP exerts favorable effects on cardiac inflammation, apoptosis, fibrosis, and mitochondrial damage both in vivo and in vitro. Further mechanistic investigations revealed that NBP blocks the interaction between Kelch-like ECH-associated protein-1 (Keap1) and Nrf2, thereby preventing the formation of the Nrf2/Keap1 complex. Discussion This study indicate that NBP alleviates DOX-induced cardiotoxicity by inhibiting Nrf2/Keap1 complex formation, highlighting its potential as a therapeutic agent for DICT and suggest that Nrf2/Keap1 may be a potential therapeutic target for the management of this condition.
Collapse
Affiliation(s)
- Yixiao Yan
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Mingzhen Fang
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Cong Zhao
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Cixi Biomedical Research Institute, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xinru Lin
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Cixi Biomedical Research Institute, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Chen Tong
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Cheng Xiang
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Ya Ran
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xuelian Wang
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Shuixin Li
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Gaozhi Chen
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Cixi Biomedical Research Institute, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Lili Fu
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
17
|
Wang J, Sun H, Peng Z, Wang SQ, Yan YQ, Luo WC, Yang RG, Bei WC, Sun LH, Yang JC. Hydroxy-Selenomethionine Supplementation During Gestation and Lactation Improve Reproduction of Sows by Enhancing the Antioxidant Capacity and Immunity Under Heat Stress Conditions. Antioxidants (Basel) 2025; 14:525. [PMID: 40427408 PMCID: PMC12108442 DOI: 10.3390/antiox14050525] [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: 02/15/2025] [Revised: 04/05/2025] [Accepted: 04/25/2025] [Indexed: 05/29/2025] Open
Abstract
The objective of this study was to determine whether hydroxy-selenomethionine (OH-SeMet) exerts better protective effects on sows against heat stress than sodium selenite (SeNa) or seleno-yeast (SeY). A total of 60 sows (Landrace × Yorkshire) were randomly allocated into the three groups and fed a base diet supplemented with SeNa, SeY, or OH-SeMet at 0.3 mg Se/kg under a heat stress condition for a reproductive cycle. Compared to SeNa or SeY, OH-SeMet could more effectively sustain offspring growth performance, as evidenced by an increased number of live-born piglets, higher litter weight at day 21, and greater litter body weight gain from days 1 to 21. OH-SeMet was more effective in supporting endogenous redox systems, as shown by enhanced levels of TXNRD and GSH and reduced levels of GSSG in the serum of sows, improved T-AOC, TXNRD, and GSH alongside decreased MDA and GSSG in the serum of piglets, and heightened T-AOC in the jejunum of piglets. Furthermore, among the two tested organic Se sources, OH-SeMet was more effective than SeY in regulating immune responses compared to SeNa. OH-SeMet reduced inflammation-related markers CRP, HP, MAP, LPS, IL-1β, IL-6, and TNF-α, some or all of which were reduced in the serum of sows and their offspring. In addition, OH-SeMet also showed reduced glucose, TG, and NEFA levels, along with elevated insulin levels in the serum of sows. Correspondingly, among the two organic forms of Se, particularly those sows fed OH-SeMet showed better gut protection for the sows' offspring, as indicated by a reduced crypt depth and increased villus height/crypt depth ratio in the duodenum, jejunum, and ileum than those fed SeNa. Specifically, compared to SeNa or SeY, OH-SeMet upregulated the expression of selenoproteins (GPX6, TXNRD3, GPX4, and SELENON), the tight junction protein (ZO-1), and host defense peptide gene (pBD1, pBD2, pBD3, NPG3, NPG4), along with downregulating levels of inflammation factor (IL-1β, IL-6 and TNF-α) and pro-apoptotic factor (P53) in the jejunum of piglets. Taken together, OH-SeMet more effectively mitigated the adverse effects induced by heat stress in sows and their offspring.
Collapse
Affiliation(s)
- Juan Wang
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Key Laboratory of Smart Farming Technology for Agricultural Animals of Ministry of Agriculture and Rural Affairs, Frontiers Science Center for Animal Breeding and Sustainable Production, College of Animal Science & Technology and College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (J.W.); (Z.P.); (S.-Q.W.); (Y.-Q.Y.); (W.-C.L.); (R.-G.Y.); (W.-C.B.)
| | - Hua Sun
- Inner Mongolia Academy of Agriculture and Animal Husbandry Science, Hohhot 010031, China;
| | - Zhe Peng
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Key Laboratory of Smart Farming Technology for Agricultural Animals of Ministry of Agriculture and Rural Affairs, Frontiers Science Center for Animal Breeding and Sustainable Production, College of Animal Science & Technology and College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (J.W.); (Z.P.); (S.-Q.W.); (Y.-Q.Y.); (W.-C.L.); (R.-G.Y.); (W.-C.B.)
| | - Shao-Qing Wang
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Key Laboratory of Smart Farming Technology for Agricultural Animals of Ministry of Agriculture and Rural Affairs, Frontiers Science Center for Animal Breeding and Sustainable Production, College of Animal Science & Technology and College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (J.W.); (Z.P.); (S.-Q.W.); (Y.-Q.Y.); (W.-C.L.); (R.-G.Y.); (W.-C.B.)
| | - Yi-Qin Yan
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Key Laboratory of Smart Farming Technology for Agricultural Animals of Ministry of Agriculture and Rural Affairs, Frontiers Science Center for Animal Breeding and Sustainable Production, College of Animal Science & Technology and College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (J.W.); (Z.P.); (S.-Q.W.); (Y.-Q.Y.); (W.-C.L.); (R.-G.Y.); (W.-C.B.)
| | - Wei-Cai Luo
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Key Laboratory of Smart Farming Technology for Agricultural Animals of Ministry of Agriculture and Rural Affairs, Frontiers Science Center for Animal Breeding and Sustainable Production, College of Animal Science & Technology and College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (J.W.); (Z.P.); (S.-Q.W.); (Y.-Q.Y.); (W.-C.L.); (R.-G.Y.); (W.-C.B.)
| | - Ren-Gui Yang
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Key Laboratory of Smart Farming Technology for Agricultural Animals of Ministry of Agriculture and Rural Affairs, Frontiers Science Center for Animal Breeding and Sustainable Production, College of Animal Science & Technology and College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (J.W.); (Z.P.); (S.-Q.W.); (Y.-Q.Y.); (W.-C.L.); (R.-G.Y.); (W.-C.B.)
| | - Wei-Cheng Bei
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Key Laboratory of Smart Farming Technology for Agricultural Animals of Ministry of Agriculture and Rural Affairs, Frontiers Science Center for Animal Breeding and Sustainable Production, College of Animal Science & Technology and College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (J.W.); (Z.P.); (S.-Q.W.); (Y.-Q.Y.); (W.-C.L.); (R.-G.Y.); (W.-C.B.)
| | - Lv-Hui Sun
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Key Laboratory of Smart Farming Technology for Agricultural Animals of Ministry of Agriculture and Rural Affairs, Frontiers Science Center for Animal Breeding and Sustainable Production, College of Animal Science & Technology and College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (J.W.); (Z.P.); (S.-Q.W.); (Y.-Q.Y.); (W.-C.L.); (R.-G.Y.); (W.-C.B.)
- Inner Mongolia Academy of Agriculture and Animal Husbandry Science, Hohhot 010031, China;
| | - Jia-Cheng Yang
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Key Laboratory of Smart Farming Technology for Agricultural Animals of Ministry of Agriculture and Rural Affairs, Frontiers Science Center for Animal Breeding and Sustainable Production, College of Animal Science & Technology and College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (J.W.); (Z.P.); (S.-Q.W.); (Y.-Q.Y.); (W.-C.L.); (R.-G.Y.); (W.-C.B.)
| |
Collapse
|
18
|
Hong KJ, Choi YJ, Kim J, Cho MC, Kim JH. Pilot study on CpG methylation of the NRF2 promoter across different ages and sexes in healthy and lung cancer prediagnostic individuals. Free Radic Biol Med 2025; 235:86-94. [PMID: 40280313 DOI: 10.1016/j.freeradbiomed.2025.04.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Revised: 04/20/2025] [Accepted: 04/22/2025] [Indexed: 04/29/2025]
Abstract
This pilot study introduces the concept of a "redox clock," an NRF2-based epigenetic clock that reflects age-related changes in oxidative stress regulation. We examined CpG methylation of the NRF2 promoter across two independent populations: 101 healthy participants (56 males, 45 females) from Gyeongsang National University Hospital (GNUH) and 150 healthy participants (111 males, 39 females) from the National Cancer Center (NCC). Methylation-sensitive HpaII restriction enzyme assays targeted three promoter regions (A, B, and C), while Illumina MethylationEPIC microarray analysis identified two specific CpG sites (cg03988329 and cg15484591) that correlated significantly with age (p < 0.01). Males, in particular, showed heightened CpG methylation in regions A and C with advancing age, alongside higher Ct values for NRF2 and HO-1 transcripts, indicating reduced gene expression. Using these methylation patterns, we developed the "redox clock" to model accelerated aging (AA). Notably, this redox clock discriminated prediagnostic lung cancer cases from healthy controls by revealing significantly greater AA in the cancer cohort, whereas established epigenetic clocks (Horvath, Hannum, and PhenoAge) did not detect this difference. These findings support CpG methylation of the NRF2 promoter, as captured by the redox clock, as a promising biomarker for biological aging and potential risk assessment for age-related diseases such as lung cancer.
Collapse
Affiliation(s)
- Ki Jae Hong
- Department of Cancer Control and Population Health, National Cancer Center Graduate School of Cancer Science and Policy, Goyang, Republic of Korea
| | - Yoon-Jung Choi
- Department of Cancer Control and Population Health, National Cancer Center Graduate School of Cancer Science and Policy, Goyang, Republic of Korea; National Cancer Control Institute, National Cancer Center, Gogyang, Republic of Korea; Center for Cancer Prevention and Detection, National Cancer Center Hospital, Goyang, Republic of Korea
| | - Jeongseon Kim
- Department of Cancer AI and Digital Health, National Cancer Center Graduate School of Cancer Science and Policy, Goyang, Republic of Korea
| | - Min-Chul Cho
- Department of Laboratory Medicine, Gyeongsang National University Hospital, Gyeongsang National University College of Medicine, Republic of Korea
| | - Jung-Hwan Kim
- Department of Pharmacology, College of Medicine, Institute of Medical Sciences, Gyeongsang National University, Jinju, Republic of Korea.
| |
Collapse
|
19
|
Wang D, Wang C, Liu H, Zhang Z, Li M, Ge X, Bi A, Gao C, Tian X, Liu K, Zhu Z. Integrated bioinformatic analysis of immune infiltration and disulfidptosis related gene subgroups in type A aortic dissection. Sci Rep 2025; 15:13719. [PMID: 40258895 PMCID: PMC12012055 DOI: 10.1038/s41598-025-98149-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 04/09/2025] [Indexed: 04/23/2025] Open
Abstract
Type A aortic dissection (TAAD) is a lethal cardiovascular disease characterized by the separation of the layers within the aortic wall. The underlying pathological mechanisms of TAAD requires further elucidation to develop effective prevention and pharmacological treatment strategies. Inflammation plays a crucial role in TAAD pathogenesis. Disulfidptosis, an emerging type of cell death, may shed light on disease mechanisms. This study investigates the role of disulfidptosis-related genes in immune infiltration in TAAD. TAAD gene expression datasets were obtained from the Gene Expression Omnibus (GEO) database. Immune cell infiltration analysis assessed immune cell dysregulation in TAAD. Differentially expressed genes (DEGs) between TAAD samples and controls were identified and intersected with known disulfidptosis-related gene sets to obtain relevant DEGs. Hub genes were identified using machine learning algorithms. A diagnostic model was constructed using Least Absolute Shrinkage and Selection Operator (LASSO) regression on 25 TAAD samples. Consensus clustering classified TAAD samples based on disulfidptosis-related gene expression. Functional enrichment analyses, including Gene Ontology (GO) terms and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses, elucidated associated biological processes and pathways. A total of 13,316 DEGs were identified, among which 11 disulfidptosis-related genes were screened: INF2, CD2AP, PDLIM1, ACTN4, MYH10, MYH9, FLNA, FLNB, TLN1, MYL6, ACTB, CAPZB, DSTN, and IQGAP1. Most of these genes exhibited lower expression levels in TAAD samples, except CAPZB, and were correlated with immune cell infiltration. Cluster-specific DEGs were found in one cluster, involving several immune response processes. Co-clustering analysis based on disulfidptosis-related genes classified TAAD samples into two clusters, with higher gene expression levels observed in cluster C2 compared to cluster C1. Three key hub genes were identified, and potential therapeutic mechanisms for TAAD were explored. Immuno-infiltration results revealed significant differences in immune profiles, with higher immunological scores and more extensive immune infiltration in TAAD. Disulfidptosis occurs in TAAD and is associated with immune cell infiltration and metabolic activity, influencing immune cell function and responses. These findings suggest that disulfidptosis may promote TAAD progression through the induction of immune responses and metabolic activities. This research provides new insights into the pathogenesis and identifies potential therapeutic targets for TAAD.
Collapse
Affiliation(s)
- Duo Wang
- Department of Cardiovascular Surgery, The Second Norman Bethune Hospital of Jilin University, Jilin University, 4026 Yatai Street, Nanguan, Changchun, Jilin, China
| | - Chengwen Wang
- Department of Cardiovascular Surgery, The Second Norman Bethune Hospital of Jilin University, Jilin University, 4026 Yatai Street, Nanguan, Changchun, Jilin, China
| | - Haoyue Liu
- Department of Cardiovascular Surgery, The Second Norman Bethune Hospital of Jilin University, Jilin University, 4026 Yatai Street, Nanguan, Changchun, Jilin, China
| | - Zhenzhu Zhang
- Graduate School, Chengde Medical University, Baoding, China
| | - Mixia Li
- Department of Cardiovascular Surgery, The Second Norman Bethune Hospital of Jilin University, Jilin University, 4026 Yatai Street, Nanguan, Changchun, Jilin, China
| | - Xinpeng Ge
- Department of Cardiovascular Surgery, The Second Norman Bethune Hospital of Jilin University, Jilin University, 4026 Yatai Street, Nanguan, Changchun, Jilin, China
| | - Anqi Bi
- Department of Cardiovascular Surgery, The Second Norman Bethune Hospital of Jilin University, Jilin University, 4026 Yatai Street, Nanguan, Changchun, Jilin, China
| | | | - Xue Tian
- Jining Public Health Medical Center, Jining, China
| | - Kexiang Liu
- Department of Cardiovascular Surgery, The Second Norman Bethune Hospital of Jilin University, Jilin University, 4026 Yatai Street, Nanguan, Changchun, Jilin, China.
| | - Zhicheng Zhu
- Department of Cardiovascular Surgery, The Second Norman Bethune Hospital of Jilin University, Jilin University, 4026 Yatai Street, Nanguan, Changchun, Jilin, China.
| |
Collapse
|
20
|
Li P, Zhu L, Song C, Wu M, Zhu X, He S, Hu B, Chen Z, Liu Z, Luo B, Liu Y, Yang X, Hu J. Triple-Functional Probiotics with Intracellularly Synthesized Selenium Nanoparticles for Colitis Therapy by Regulating the Macrophage Phenotype and Modulating Gut Microbiota. ACS NANO 2025; 19:14213-14232. [PMID: 40192063 DOI: 10.1021/acsnano.5c00574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2025]
Abstract
The dysregulated macrophage phenotype, as the main cause of colitis, not only enhanced oxidative stress to exacerbate inflammatory responses but was closely related with gut microbial dysbiosis. It was needed to simultaneously address the three issues for the effective treatment of colitis, but it was not satisfied. Here, we developed "three-birds-one-stone" probiotics, named Se@EcN-C2/A2, for colitis treatment. Escherichia coli Nissle 1917 (EcN), a clinically approved probiotic, was used to intracellularly synthesize selenium (Se) nanoparticles by biomineralization, giving Se@EcN. Coating glycol chitosan and sodium alginate on the surface of Se@EcN (Se@EcN-C2/A2) endowed probiotics with high resistance to the harsh gastrointestinal tract environment and strong adhesion and targeting ability to the inflamed site of the colon to facilitate the uptake by M1 macrophages. Se@EcN-C2/A2 was metabolized to SeCys2 and MetSeCys to be involved in the synthesis of GPX2 and TXNRD1, which led to reaction oxygen species clearance to inhibit Toll-like receptor and nuclear factor κB signaling pathways to suppress inflammatory response and polarize M1 macrophages to M2 phenotypes by activating PI3K/AKT signaling pathways. In DSS-induced colitis mice, Se@EcN-C2/A2 exerted satisfactory therapeutic and prophylactic effects, including scavenging oxidative stress and regulating macrophage phenotypes to suppress inflammatory response and restore gut barrier functions. Moreover, the living probiotic EcN in the colon effectively regulated microbial dysbiosis by decreasing the abundance of Escherichia-Shigella and increasing the abundance of Lactobacillus and Bifidobacterium.
Collapse
Affiliation(s)
- Puze Li
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Lichong Zhu
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Cheng Song
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Meichan Wu
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Xuan Zhu
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Suting He
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Bo Hu
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Zehao Chen
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Zhi Liu
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Ban Luo
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yan Liu
- Department of Gastroenterology, Wuhan Eighth Hospital, Wuhan 430010, China
| | - Xiangliang Yang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Jun Hu
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- Hubei Jiangxia Laboratory, Wuhan 430200, China
| |
Collapse
|
21
|
Polo-Montalvo A, Gómez-Cerezo N, Cicuéndez M, González B, Izquierdo-Barba I, Arcos D. Osteogenic and Antibacterial Response of Levofloxacin-Loaded Mesoporous Nanoparticles Functionalized with N-Acetylcysteine. Pharmaceutics 2025; 17:519. [PMID: 40284514 PMCID: PMC12030205 DOI: 10.3390/pharmaceutics17040519] [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: 02/28/2025] [Revised: 04/01/2025] [Accepted: 04/11/2025] [Indexed: 04/29/2025] Open
Abstract
Background/Objectives: Bone infection is one of the most prevalent complications in orthopedic surgery. This pathology is mostly due to bacterial pathogens, among which S. aureus stands out. The formation of a bacterial biofilm makes systemic treatment with antibiotics ineffective. Herein we propose a nanosystem composed of mesoporous bioactive glass nanoparticles (MBGN) loaded with levofloxacin and functionalized with N-acetylcysteine (NAC), aiming to offer an alternative to current treatments. These nanoparticles would present antibacterial activity able to disintegrate the biofilm and regenerate the peri-implantar osseous tissue. Methods: MBGN of composition 82.5 SiO2-17.5 CaO have been synthesized, loaded with levofloxacin, and functionalized with NAC (MBGN-L-NAC). The antimicrobial activity against mature S. aureus biofilms and bioactivity of the nanosystem have been evaluated, as well as its biocompatibility and ability to promote murine pre-osteoblastic MC3T3-E1 differentiation. Results: MBGNs exhibited high surface areas and radial mesoporosity, allowing up to 23.1% (% w/w) of levofloxacin loading. NAC was covalently bound keeping the mucolytic thiol group, SH, available. NAC and levofloxacin combination enhances the activity against S. aureus by disrupting mature biofilm integrity. This nanosystem was biocompatible with pre-osteoblasts, enhanced their differentiation towards a mature osteoblast phenotype, and promoted bio-mimetic mineralization under in vitro conditions. MBGN-L-NAC nanoparticles induced greater osteogenic response of osteoprogenitor cells through increased alkaline phosphatase expression, increased mineralization, and stimulation of pre-osteoblast nodule formation. Conclusions: MBGN-L-NAC exhibits a more efficient antibacterial activity due to the biofilm disaggregation exerted by NAC, which also contributes to enhance the osteoinductive properties of MBGNs, providing a potential alternative to conventional strategies for the management of bone infections.
Collapse
Affiliation(s)
- Alberto Polo-Montalvo
- Departamento Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense de Madrid, Plaza Ramón y Cajal s/n, 28040 Madrid, Spain; (A.P.-M.); (N.G.-C.); (M.C.); (B.G.)
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
| | - Natividad Gómez-Cerezo
- Departamento Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense de Madrid, Plaza Ramón y Cajal s/n, 28040 Madrid, Spain; (A.P.-M.); (N.G.-C.); (M.C.); (B.G.)
- Instituto de Investigación Sanitaria, Hospital 12 de Octubre i+12, 28040 Madrid, Spain
| | - Mónica Cicuéndez
- Departamento Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense de Madrid, Plaza Ramón y Cajal s/n, 28040 Madrid, Spain; (A.P.-M.); (N.G.-C.); (M.C.); (B.G.)
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
| | - Blanca González
- Departamento Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense de Madrid, Plaza Ramón y Cajal s/n, 28040 Madrid, Spain; (A.P.-M.); (N.G.-C.); (M.C.); (B.G.)
- Instituto de Investigación Sanitaria, Hospital 12 de Octubre i+12, 28040 Madrid, Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina—CIBER-BBN, 28040 Madrid, Spain
| | - Isabel Izquierdo-Barba
- Departamento Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense de Madrid, Plaza Ramón y Cajal s/n, 28040 Madrid, Spain; (A.P.-M.); (N.G.-C.); (M.C.); (B.G.)
- Instituto de Investigación Sanitaria, Hospital 12 de Octubre i+12, 28040 Madrid, Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina—CIBER-BBN, 28040 Madrid, Spain
| | - Daniel Arcos
- Departamento Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense de Madrid, Plaza Ramón y Cajal s/n, 28040 Madrid, Spain; (A.P.-M.); (N.G.-C.); (M.C.); (B.G.)
- Instituto de Investigación Sanitaria, Hospital 12 de Octubre i+12, 28040 Madrid, Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina—CIBER-BBN, 28040 Madrid, Spain
| |
Collapse
|
22
|
Zhang X, Han S, Zeng Z, Dai J, Jia Y. Selenium-Binding Protein 1-Deficient Dendritic Cells Protect Mice from Sepsis by Increased Treg/Th17. Antioxidants (Basel) 2025; 14:468. [PMID: 40298842 PMCID: PMC12024190 DOI: 10.3390/antiox14040468] [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: 03/12/2025] [Revised: 04/08/2025] [Accepted: 04/10/2025] [Indexed: 04/30/2025] Open
Abstract
Selenium-binding protein 1 (SELENBP1) has been implicated in cancer development, neurological disorders, tissue injury, metabolic regulation, and cell differentiation. Sepsis is characterized prominently by immunological dysregulation and severe organ damage. However, whether SELENBP1 improves sepsis by regulating immune cell activity remains unknown. Here, we detected an elevation of SELENBP1 levels in the blood of sepsis patients and in the livers of septic mice. Significantly, SELENBP1 knockout (KO) prolonged survival in septic mice. This phenomenon was accompanied by decreased liver damage, reduced inflammation levels, and an increased regulatory T cell/T helper 17 cell (Treg/Th17) ratio in the spleen. Additionally, SELENBP1 deficiency induced a redox imbalance and inhibited dendritic cell (DC) maturation, resulting in a tolerogenic DC (tolDC) phenotype and an increase in the Treg/Th17 ratio. Furthermore, SELENBP1-KO mature DCs (mDCs) alleviated liver injury by increasing the Treg/Th17 ratio in the spleen, thus improving the survival of septic mice. These findings indicate that SELENBP1 is involved in sepsis by regulating DC immune activity, which might provide a potential way for sepsis treatment.
Collapse
Affiliation(s)
- Xin Zhang
- Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province, Cellular Immunotherapy Engineering Research Center of Guizhou Province, School of Basic Medical Sciences/School of Biology and Engineering (School of Modern Industry for Health and Medicine), Guizhou Medical University, Guiyang 550025, China; (X.Z.); (S.H.); (Z.Z.)
| | - Shuang Han
- Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province, Cellular Immunotherapy Engineering Research Center of Guizhou Province, School of Basic Medical Sciences/School of Biology and Engineering (School of Modern Industry for Health and Medicine), Guizhou Medical University, Guiyang 550025, China; (X.Z.); (S.H.); (Z.Z.)
| | - Zhu Zeng
- Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province, Cellular Immunotherapy Engineering Research Center of Guizhou Province, School of Basic Medical Sciences/School of Biology and Engineering (School of Modern Industry for Health and Medicine), Guizhou Medical University, Guiyang 550025, China; (X.Z.); (S.H.); (Z.Z.)
- Immune Cells and Antibody Engineering Research Center of Guizhou Province, Key Laboratory of Biology and Medical Engineering, Guizhou Medical University, Guiyang 550025, China
| | - Jie Dai
- Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province, Cellular Immunotherapy Engineering Research Center of Guizhou Province, School of Basic Medical Sciences/School of Biology and Engineering (School of Modern Industry for Health and Medicine), Guizhou Medical University, Guiyang 550025, China; (X.Z.); (S.H.); (Z.Z.)
| | - Yi Jia
- Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province, Cellular Immunotherapy Engineering Research Center of Guizhou Province, School of Basic Medical Sciences/School of Biology and Engineering (School of Modern Industry for Health and Medicine), Guizhou Medical University, Guiyang 550025, China; (X.Z.); (S.H.); (Z.Z.)
- Immune Cells and Antibody Engineering Research Center of Guizhou Province, Key Laboratory of Biology and Medical Engineering, Guizhou Medical University, Guiyang 550025, China
| |
Collapse
|
23
|
Yu P, Bo L. Effects of Sevoflurane and Propofol During Mechanical Ventilation: A Meta-analysis of Randomized Controlled Trials. J Perianesth Nurs 2025:S1089-9472(24)00562-8. [PMID: 40202471 DOI: 10.1016/j.jopan.2024.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 11/08/2024] [Accepted: 12/05/2024] [Indexed: 04/10/2025]
Abstract
PURPOSE To compare the inflammatory response, hemodynamic stability, and postoperative recovery of sevoflurane versus propofol after mechanical ventilation during surgery, to provide references for rational anesthesia utility in clinical practice by meta-analysis. DESIGN Systematic review and meta-analysis. METHODS Pubmed, Web of Science, Cochrane Library, Wanfang Data, China National Knowledge Infrastructure, and Chinese BioMedical Literature Database were searched by computer for trials on the anesthetic effects of sevoflurane and propofol after mechanical ventilation during surgery. A random- or fixed-effects model was applied to analyze the clinical indicators and adverse impact based on heterogeneity. FINDINGS As of April 21, 2023, 912 articles were retrieved, and 36 eligible articles were finally identified after screening, covering 2,691 surgical patients for meta-analysis. The combined results exhibited that the level of tumor necrosis factor (TNF)-α in alveolar lavage was significantly different between the 2 groups (-0.94, 95% confidence interval [CI]: -1.82 to -0.05, P = .038). Compared with sevoflurane, propofol significantly increased TNF-α levels in alveolar lavage. In plasma or serum, propofol significantly increased the levels of interleukin-10 (-0.73, 95% CI: -1.36 to -0.10, P = .023) and TNF-α (-0.65, 95% CI: -1.21 to -0.09, P = .022). CONCLUSIONS At alveolar lavage and serum or plasma levels of inflammatory factors, the proinflammatory factor TNF-α was significantly lower in the sevoflurane group than in the propofol group. This indicates that sevoflurane has a certain role in alleviating local and systemic lung inflammation. However, more randomized controlled studies are warranted in the future to confirm whether there is a difference in hemodynamic stability and postoperative recovery of patients.
Collapse
Affiliation(s)
- Peixia Yu
- Department of Anaesthesiology, The Third Hospital of Hebei Medical University, Shijiazhuang, Heibei, China
| | - Lijun Bo
- Department of Anaesthesiology, The Second Hospital of Hebei Medical University, Shijiazhuang, Heibei, China.
| |
Collapse
|
24
|
Li N, Pang Y, Xu J, Elango J, Wu W. Immunomodulatory Effects of Symplectoteuthis oualaniensis Protamine and Its PEG Derivative on Macrophages: Involvement of PI3K/Akt Signaling, Redox Regulation, and Cell Cycle Modulation. Antioxidants (Basel) 2025; 14:437. [PMID: 40298789 PMCID: PMC12024133 DOI: 10.3390/antiox14040437] [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: 03/08/2025] [Revised: 04/02/2025] [Accepted: 04/03/2025] [Indexed: 04/30/2025] Open
Abstract
Protamine is a promising marine-derived bioactive compound that is highly arginine-rich and has demonstrated unique advantages in medical and biological research. This study, for the first time, investigates the molecular mechanisms underlying the immunomodulatory effects of Salmon Protamine Sulfate (SPS), Symplectoteuthis oualaniensis Protamine (SOP), and its polyethylene glycol (PEG) derivative (SOP-PEG) on RAW264.7 macrophages. The results demonstrate that both SOP and SOP-PEG significantly enhance the proliferation of RAW264.7 cells by promoting the secretion of pro-inflammatory cytokines and nitric oxide (NO), increasing ROS production, and improving antioxidant capacity, in comparison to SPS. Elevated ROS levels play a crucial role in enhancing macrophage immune activity, while the enhanced antioxidant defense mechanisms help maintain redox homeostasis and protect against oxidative stress-induced cellular damage. A Western blot analysis reveals that SOP and SOP-PEG notably regulate the expression of key proteins associated with the PI3K/Akt signaling pathway and anti-apoptotic mechanisms. Furthermore, a flow cytometry analysis indicates a significant increase in the G2/M-phase cell population in the treatment groups, which is corroborated by Western blot data showing alterations in critical regulatory proteins. Notably, SOP-PEG exhibits the strongest effects in regulating macrophage immune activity, which can be attributed to the enhanced stability and prolonged bioactivity resulting from the PEGylation of SOP. This comprehensive study reveals how SOP and SOP-PEG enhance macrophage immune function through multiple mechanisms, including PI3K/Akt activation, redox regulation, and cell cycle modulation. It provides valuable insights and a theoretical foundation for their potential applications in immunotherapy and immune regulation.
Collapse
Affiliation(s)
- Na Li
- Department of Marine Pharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China; (N.L.); (Y.P.); (J.X.)
| | - Yida Pang
- Department of Marine Pharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China; (N.L.); (Y.P.); (J.X.)
| | - Jiren Xu
- Department of Marine Pharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China; (N.L.); (Y.P.); (J.X.)
| | - Jeevithan Elango
- Department of Marine Pharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China; (N.L.); (Y.P.); (J.X.)
- Department of Biomaterials Engineering, Faculty of Health Sciences, UCAM-Universidad Católica San Antonio de Murcia, Guadalupe, 30107 Murcia, Spain
- Center of Molecular Medicine and Diagnostics (COMManD), Department of Biochemistry, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 600077, India
| | - Wenhui Wu
- Department of Marine Pharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China; (N.L.); (Y.P.); (J.X.)
- Marine Biomedical Science and Technology Innovation Platform of Lin-gang Special Area, Shanghai 201306, China
- Putuo Branch of International Combined Research Center for Marine Biological Sciences, Zhoushan 316104, China
| |
Collapse
|
25
|
Luo W, Xu C, Li L, Ji Y, Wang Y, Li Y, Ye Y. Perfluoropentane-based oxygen-loaded nanodroplets reduce microglial activation through metabolic reprogramming. Neural Regen Res 2025; 20:1178-1191. [PMID: 38989955 PMCID: PMC11438333 DOI: 10.4103/nrr.nrr-d-23-01299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 02/05/2024] [Indexed: 07/12/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202504000-00032/figure1/v/2024-07-06T104127Z/r/image-tiff Microglia, the primary immune cells within the brain, have gained recognition as a promising therapeutic target for managing neurodegenerative diseases within the central nervous system, including Parkinson's disease. Nanoscale perfluorocarbon droplets have been reported to not only possess a high oxygen-carrying capacity, but also exhibit remarkable anti-inflammatory properties. However, the role of perfluoropentane in microglia-mediated central inflammatory reactions remains poorly understood. In this study, we developed perfluoropentane-based oxygen-loaded nanodroplets (PFP-OLNDs) and found that pretreatment with these droplets suppressed the lipopolysaccharide-induced activation of M1-type microglia in vitro and in vivo, and suppressed microglial activation in a mouse model of Parkinson's disease. Microglial suppression led to a reduction in the inflammatory response, oxidative stress, and cell migration capacity in vitro. Consequently, the neurotoxic effects were mitigated, which alleviated neuronal degeneration. Additionally, ultrahigh-performance liquid chromatography-tandem mass spectrometry showed that the anti-inflammatory effects of PFP-OLNDs mainly resulted from the modulation of microglial metabolic reprogramming. We further showed that PFP-OLNDs regulated microglial metabolic reprogramming through the AKT-mTOR-HIF-1α pathway. Collectively, our findings suggest that the novel PFP-OLNDs constructed in this study alleviate microglia-mediated central inflammatory reactions through metabolic reprogramming.
Collapse
Affiliation(s)
- Wanxian Luo
- Department of Medicine Ultrasonics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Chuanhui Xu
- Institute of Neuroscience, Department of Neurosurgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Linxi Li
- Institute of Neuroscience, Department of Neurosurgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Yunxiang Ji
- Institute of Neuroscience, Department of Neurosurgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Yezhong Wang
- Institute of Neuroscience, Department of Neurosurgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Yingjia Li
- Department of Medicine Ultrasonics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Yongyi Ye
- Institute of Neuroscience, Department of Neurosurgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong Province, China
| |
Collapse
|
26
|
Ohtsu H. - Invited Review - Challenges and constraints in the sustainability of poultry farming in Japan. Anim Biosci 2025; 38:818-828. [PMID: 39999794 PMCID: PMC11969155 DOI: 10.5713/ab.24.0675] [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: 09/27/2024] [Revised: 11/12/2024] [Accepted: 01/22/2025] [Indexed: 02/27/2025] Open
Abstract
Poultry products such as chicken meat and eggs are among the most common and popular animal products in Japan. Recently, many chickens, such as broilers and layers, have been raised and their related product consumption has increased. However, the number of farms decreased, which is one of the major challenges faced by the Japanese poultry industry. Similar to that in other countries, high-pathogenicity avian influenza (HPAI) outbreaks negatively affected the distribution of poultry products. Low feed selfsufficiency in Japan is also a serious problem because the prices of diets and products are affected by the situation in foreign countries. Rice is a domestic ingredient of the poultry diet in Japan, and recently, its utilization has increased; however, concerns remain. Global warming likely affects the poultry industry in Japan negatively. The objective of this review is to illustrate the recent situation of the Japanese poultry industry, including 1) an overview; 2) the situation of influence of HPAI; 3) situation of ingredients for poultry diet; 4) utilization of rice in poultry diet; 5) heat stress in poultry. Overall, investigation of the effects of heat stress on physiology, such as the biological defense system, and its prevention, should be continued to prevent future decreases in productivity in the Japanese poultry industry.
Collapse
Affiliation(s)
- Haruhiko Ohtsu
- Institute of Livestock and Grassland Science, NARO, Tsukuba,
Japan
| |
Collapse
|
27
|
She Y, Wu P, Wan W, Liu H, Liu R, Wang T, Wang M, Shen L, Yang Y, Huang X, Zhang X, Tian Y, Zhang K. Polysaccharides, proteins and DNA based stimulus responsive hydrogels promoting wound healing and repair: A review. Int J Biol Macromol 2025; 304:140961. [PMID: 39952504 DOI: 10.1016/j.ijbiomac.2025.140961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 02/08/2025] [Accepted: 02/11/2025] [Indexed: 02/17/2025]
Abstract
The healing of various wounds remains a serious challenge in the medical field, hydrogel has high hydrophilicity and biocompatibility due to its unique network structure, which shows a strong advantage in the field of wound healing. Stimulus responsive hydrogels are particularly effective,which can control the material properties according to the external stimulus source, and provide more targeted treatment for different wounds. Here, we review physiological mechanisms of wound healing and the relationship between polysaccharides, proteins and DNA based stimulus responsive hydrogels and wound healing, materials commonly used of polysaccharides, proteins and DNA based stimulus responsive hydrogels, mechanisms of stimulus responsive hydrogels formation and network structure types, common properties of polysaccharides, proteins and DNA based stimulus responsive hydrogels for promoting wound healing and discuss their applications in medicine. Finally, the limitations and application prospects of polysaccharides, proteins and DNA based stimulus responsive hydrogels were discussed and evaluated. The review focuses on the biomedical use of polysaccharides, proteins and DNA based stimulus responsive hydrogels in wound healing and repair, and provides insights for the development of clinical related materials.
Collapse
Affiliation(s)
- Yumo She
- Department of Gastroenterology, Endoscopic Center, Shengjing Hospital of China Medical University, China
| | - Peng Wu
- Department of Gastroenterology, Endoscopic Center, Shengjing Hospital of China Medical University, China
| | - Wenyu Wan
- Key Laboratory of Immunodermatology, Ministry of Education, Department of Dermatology, The First Hospital of China Medical University, China; Key Laboratory of Immunodermatology, National Health Commission of the People's Republic of China, The First Hospital of China Medical University, China; National and Local Joint Engineering Research Center of Immunodermatological Theranostics, The First Hospital of China Medical University, China
| | - He Liu
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang 110169, China
| | - Ruonan Liu
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang 110169, China
| | - Tingting Wang
- Department of Gastroenterology, Endoscopic Center, Shengjing Hospital of China Medical University, China
| | - Mengyao Wang
- Department of Gastroenterology, Endoscopic Center, Shengjing Hospital of China Medical University, China
| | - Lufan Shen
- Department of Gastroenterology, Endoscopic Center, Shengjing Hospital of China Medical University, China
| | - Yuanyuan Yang
- Department of Gastroenterology, Endoscopic Center, Shengjing Hospital of China Medical University, China
| | - Xingyong Huang
- Department of Gastroenterology, Endoscopic Center, Shengjing Hospital of China Medical University, China
| | - Xiaoyue Zhang
- Department of Gastroenterology, Endoscopic Center, Shengjing Hospital of China Medical University, China
| | - Ye Tian
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang 110169, China; Foshan Graduate School of Innovation, Northeastern University, Foshan 528300, China.
| | - Kai Zhang
- Department of Gastroenterology, Endoscopic Center, Shengjing Hospital of China Medical University, China; Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, China.
| |
Collapse
|
28
|
Cotzomi-Ortega I, Rosowski EE, Wang X, Sanchez-Zamora YI, Lopez-Torres JM, Sanchez-Orellana G, Han R, Vásquez-Martínez G, Andrade GM, Ballash G, Cortado H, Li B, Ali Y, Rascon R, Robledo-Avila F, Partida-Sanchez S, Pérez-Campos E, Olofsson-Sahl P, Zepeda-Orozco D, Spencer JD, Becknell B, Ruiz-Rosado JDD. Neutrophil NADPH oxidase promotes bacterial eradication and regulates NF-κB-Mediated inflammation via NRF2 signaling during urinary tract infections. Mucosal Immunol 2025; 18:402-417. [PMID: 39710133 PMCID: PMC11993340 DOI: 10.1016/j.mucimm.2024.12.010] [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: 08/05/2024] [Revised: 11/18/2024] [Accepted: 12/15/2024] [Indexed: 12/24/2024]
Abstract
The precise role of neutrophil-derived reactive oxygen species (ROS) in combating bacterial uropathogens during urinary tract infections (UTI) remains largely unexplored. In this study, we elucidate the antimicrobial significance of NADPH oxidase 2 (NOX2)-derived ROS, as opposed to mitochondrial ROS, in facilitating neutrophil-mediated eradication of uropathogenic Escherichia coli (UPEC), the primary causative agent of UTI. Furthermore, NOX2-derived ROS regulate NF-κB-mediated inflammatory responses in neutrophils against UPEC by inducing the release of nuclear factor erythroid 2-related factor 2 (Nrf2) from its inhibitor, Kelch-like ECH-associated protein 1 (Keap1). Consistently, the absence of NOX2 (Cybb-/-) in mice led to uncontrolled bacterial infection associated with increased NF-κB signaling, heightened neutrophilic inflammation, and increased bladder pathology during cystitis. These findings underscore a dual role for neutrophil NOX2 in both eradicating UPEC and mitigating neutrophil-mediated inflammation in the urinary tract, revealing a previously unrecognized effector and regulatory mechanism in the control of UTI.
Collapse
Affiliation(s)
- Israel Cotzomi-Ortega
- Kidney and Urinary Tract Center, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH 43205, USA
| | - Emily E Rosowski
- Department of Biological Sciences, Clemson University, Clemson, SC 29634, USA; Eukaryotic Pathogens Innovation Center, Clemson University, Clemson, SC 29634, USA
| | - Xin Wang
- Kidney and Urinary Tract Center, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH 43205, USA
| | - Yuriko I Sanchez-Zamora
- Kidney and Urinary Tract Center, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH 43205, USA
| | - Jeimy M Lopez-Torres
- Kidney and Urinary Tract Center, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH 43205, USA
| | - Gamaliel Sanchez-Orellana
- Kidney and Urinary Tract Center, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH 43205, USA
| | - Rachel Han
- Indiana University School of Medicine, Riley Hospital for Children at Indiana University Health, Division of Neonatology, Indianapolis, IN, USA
| | - Gabriela Vásquez-Martínez
- Kidney and Urinary Tract Center, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH 43205, USA
| | - Gabriel Mayoral Andrade
- Kidney and Urinary Tract Center, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH 43205, USA; Center for Research in Medicine, National Autonomous University of Mexico-Autonomous University Benito Juárez of Oaxaca (UNAM-UABJO), Faculty of Medicine and Surgery, Autonomous University Benito Juárez of Oaxaca, Oaxaca 68020, Mexico
| | - Gregory Ballash
- Department of Veterinary Preventive Medicine, The Ohio State University, Columbus, OH, USA
| | - Hanna Cortado
- Kidney and Urinary Tract Center, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH 43205, USA
| | - Birong Li
- Kidney and Urinary Tract Center, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH 43205, USA
| | - Yusuf Ali
- Kidney and Urinary Tract Center, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH 43205, USA
| | - Raul Rascon
- Center for Microbial Pathogenesis, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH 43205, USA
| | - Frank Robledo-Avila
- Center for Microbial Pathogenesis, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH 43205, USA
| | - Santiago Partida-Sanchez
- Center for Microbial Pathogenesis, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH 43205, USA
| | | | - Peter Olofsson-Sahl
- Pronoxis AB, SE-461 33 Trollhättan, Sweden; University West, Department of Health Sciences, SE-461 86 Trollhättan, Sweden
| | - Diana Zepeda-Orozco
- Kidney and Urinary Tract Center, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH 43205, USA; Division of Nephrology and Hypertension, Nationwide Children's Hospital, Columbus, OH 43215, USA
| | - John David Spencer
- Kidney and Urinary Tract Center, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH 43205, USA; Division of Nephrology and Hypertension, Nationwide Children's Hospital, Columbus, OH 43215, USA
| | - Brian Becknell
- Kidney and Urinary Tract Center, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH 43205, USA; Division of Nephrology and Hypertension, Nationwide Children's Hospital, Columbus, OH 43215, USA.
| | - Juan de Dios Ruiz-Rosado
- Kidney and Urinary Tract Center, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH 43205, USA; Division of Nephrology and Hypertension, Nationwide Children's Hospital, Columbus, OH 43215, USA.
| |
Collapse
|
29
|
Li S, Yuan H, Yang XZ, Xu X, Yu W, Wu Y, Yao S, Xie J, He W, Guo Z, Chen Y. Synergistic Antitumor Immunotherapy via Mitochondria Regulation in Macrophages and Tumor Cells by an Iridium Photosensitizer. ACS CENTRAL SCIENCE 2025; 11:441-451. [PMID: 40161955 PMCID: PMC11950858 DOI: 10.1021/acscentsci.4c02156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 02/13/2025] [Accepted: 03/03/2025] [Indexed: 04/02/2025]
Abstract
Mitochondrial targeting has emerged as an attractive method for antitumor treatment. However, most of the mitochondria targeted drugs focused on inhibiting tumor cells, while their potential for activation of immune responses in the tumor microenvironment has rarely been described. In this study, we report a photosensitive iridium complex MitoIrL2, which enabled the simultaneous mitochondrial modulation of macrophages and tumor cells to achieve synergistic antitumor immunity. The adjustment of the mitochondrial respiratory chain, HIF-1α, and the NF-κB pathway in macrophages drove the metabolic reprogramming from oxidative phosphorylation (OXPHOS) to glycolysis, converting protumor M2 into the antitumor M1 phenotype. Downregulated expression of immunosuppressive checkpoint SIRPα has also been observed on macrophages. Meanwhile, the mitochondrial targeting MitoIrL2 enhanced the immunogenic cell death of tumor cells and reversed the immunosuppressive tumor microenvironment, which activated the systemic immune response and established long-term immune memory in vivo. This work illustrates a promising strategy to simultaneously regulate macrophages toward the antitumor phenotype and enhance immunogenic cell death in tumor cells for synergistic antitumor immunotherapy.
Collapse
Affiliation(s)
- Shumeng Li
- State
Key Laboratory of Coordination Chemistry, School of Chemistry and
Chemical Engineering, Chemistry and Biomedicine Innovation Center
(ChemBIC), ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing 210023, Jiangsu, P.R. China
| | - Hao Yuan
- State
Key Laboratory of Coordination Chemistry, School of Chemistry and
Chemical Engineering, Chemistry and Biomedicine Innovation Center
(ChemBIC), ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing 210023, Jiangsu, P.R. China
| | - Xiu-Zhi Yang
- State
Key Laboratory of Coordination Chemistry, School of Chemistry and
Chemical Engineering, Chemistry and Biomedicine Innovation Center
(ChemBIC), ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing 210023, Jiangsu, P.R. China
| | - Xinyu Xu
- State
Key Laboratory of Coordination Chemistry, School of Chemistry and
Chemical Engineering, Chemistry and Biomedicine Innovation Center
(ChemBIC), ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing 210023, Jiangsu, P.R. China
| | - Wenhao Yu
- State
Key Laboratory of Coordination Chemistry, School of Chemistry and
Chemical Engineering, Chemistry and Biomedicine Innovation Center
(ChemBIC), ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing 210023, Jiangsu, P.R. China
| | - Yanping Wu
- State
Key Laboratory of Coordination Chemistry, School of Chemistry and
Chemical Engineering, Chemistry and Biomedicine Innovation Center
(ChemBIC), ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing 210023, Jiangsu, P.R. China
| | - Shankun Yao
- State
Key Laboratory of Coordination Chemistry, School of Chemistry and
Chemical Engineering, Chemistry and Biomedicine Innovation Center
(ChemBIC), ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing 210023, Jiangsu, P.R. China
| | - Jin Xie
- State
Key Laboratory of Coordination Chemistry, School of Chemistry and
Chemical Engineering, Chemistry and Biomedicine Innovation Center
(ChemBIC), ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing 210023, Jiangsu, P.R. China
| | - Weijiang He
- State
Key Laboratory of Coordination Chemistry, School of Chemistry and
Chemical Engineering, Chemistry and Biomedicine Innovation Center
(ChemBIC), ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing 210023, Jiangsu, P.R. China
| | - Zijian Guo
- State
Key Laboratory of Coordination Chemistry, School of Chemistry and
Chemical Engineering, Chemistry and Biomedicine Innovation Center
(ChemBIC), ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing 210023, Jiangsu, P.R. China
| | - Yuncong Chen
- State
Key Laboratory of Coordination Chemistry, School of Chemistry and
Chemical Engineering, Chemistry and Biomedicine Innovation Center
(ChemBIC), ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing 210023, Jiangsu, P.R. China
- Department
of Cardiothoracic Surgery, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing 210008, Jiangsu, P.R. China
| |
Collapse
|
30
|
Li G, Wang Z, Guo Y, Ni C, Gao Y, Xu K, Xiao T, Shi X, Shen M. Copper-doped layered double hydroxides co-deliver proteins/drugs for cascaded chemodynamic/immunotherapy via dual regulation of tumor metabolism. Acta Biomater 2025; 195:350-362. [PMID: 39921184 DOI: 10.1016/j.actbio.2025.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 01/17/2025] [Accepted: 02/04/2025] [Indexed: 02/10/2025]
Abstract
We report here a Cu2+-doped layered double hydroxide (LDH) nanoplatform to load both monocarboxylate transporter inhibitor diclofenac (DC) and lactate oxidase (LOX) for dual modulation of tumor lactate and redox metabolisms to activate immunotherapy through enhanced Cu-mediated chemodynamic therapy (CDT) of tumors. The formed LDH-DC-LOX nanoparticles with a diameter of 55 nm are stable, can be effectively taken up by cancer cells to regulate lactate through both LOX-mediated catalytic conversion and DC-enabled inhibition of extracellular efflux of lactate, and can exert redox metabolism through CDT via Cu2+-mediated glutathione (GSH) depletion and Fenton-like reaction with hydrogen peroxide (H2O2) that can be further accumulated via LOX-mediated catalysis. The major advantage of the developed LDH-DC-LOX nanoparticles lies in the therapeutic synergy and cascade that can be achieved through the loaded DC and LOX for enhanced tumor lactate metabolism regulation, for enhanced Cu-mediated CDT and redox metabolism regulation, and for activation of immunotherapy that can further enhances the Cu2+-mediated CDT effect. The developed LDH nanoplatform demonstrated here for effective murine breast tumor treatment provides a new paradigm for dual regulation of lactate and redox metabolisms that may enable synergistic and cascaded combination therapy of different cancer types. STATEMENT OF SIGNIFICANCE: Targeting the tumor microenvironment (TME) to alter tumor metabolic pathways represents a promising strategy for next-generation cancer therapy. Herein, a copper-doped layered double hydroxide (LDH) nanoplatform is developed to co-deliver both diclofenac (DC) and lactate oxidase (LOX) to tumor cells for efficient dual regulation of tumor lactate and redox metabolisms, resulting in synergistic and cascaded chemodynamic therapy/immunotherapy of breast tumors. The developed LDH-DC-LOX nanoparticles can release Cu2+ and DC under an acidic TME, and can act in synergy to reduce TME lactate and generate H2O2, thus modulating redox metabolism and activating anticancer immunotherapy. The secreted cytokine IFN-γ after activation of antitumor immune responses can further mediate enhanced chemodynamic therapy effect through downregulation of cystine/glutamate transporter SLC7A11 to suppress GSH synthesis.
Collapse
Affiliation(s)
- Gaoming Li
- State Key Laboratory of Advanced Fiber Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Zhiqiang Wang
- State Key Laboratory of Advanced Fiber Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Yunqi Guo
- State Key Laboratory of Advanced Fiber Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Cheng Ni
- State Key Laboratory of Advanced Fiber Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Yue Gao
- State Key Laboratory of Advanced Fiber Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Kaibing Xu
- Research Center for Analysis and Measurement, Donghua University, Shanghai 201620, China
| | - Tingting Xiao
- Institute of Frontier Medical Technology, College of Chemistry and Chemical Engineering, Shanghai University of Engineering Science, Shanghai 201620, China
| | - Xiangyang Shi
- State Key Laboratory of Advanced Fiber Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China; CQM-Centro de Química da Madeira, Universidade da Madeira, Campus Universitário da Penteada, Funchal 9020-105, Portugal.
| | - Mingwu Shen
- State Key Laboratory of Advanced Fiber Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China.
| |
Collapse
|
31
|
Ye J, Qin Y, Liu H, Xiong H, Zhang H, Shen H, Zeng F, Shi C, Zhou Z. Inhibiting Neutrophil Extracellular Trap Formation through Iron Regulation for Enhanced Cancer Immunotherapy. ACS NANO 2025; 19:9167-9181. [PMID: 40011227 DOI: 10.1021/acsnano.4c18555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/28/2025]
Abstract
Iron metabolism of neutrophils plays a vital role in neutrophil extracellular trap (NET) formation, which presents as one of the major hurdles to the immune response in the tumor microenvironment. Here, we developed a peptide-drug conjugate (PDC)-based transformable iron nanochelator (TIN) equipped with the ability to regulate the iron metabolism of neutrophils, endowing inhibition of NET formation and the ensuing immunosuppression functions. The TIN could expose the iron-binding motifs through neutrophil elastase-mediated morphological transformation from nanoparticles to β-sheet nanofibers, which further evolve into stable α-helix nanofibers after chelation with iron(II) ions. This process enables a highly specific regulation of iron(II) ions of neutrophils, which turns into an efficient way of inhibiting NET formation and improving the immune response. Furthermore, the TIN showed an improved therapeutic effect in combination with protein arginine deiminase 4 inhibitors and synergistically boosted the anti-PD-L1 treatment. This study designates an iron-regulation strategy to inhibit NET formation, which provides an alternative approach to immune modulation from the perspective of targeting the iron metabolism of neutrophils in cancer immunotherapy.
Collapse
Affiliation(s)
- Jinmin Ye
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory & Center for Molecular Imaging and Translational Medicine, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Yatong Qin
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory & Center for Molecular Imaging and Translational Medicine, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Hui Liu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory & Center for Molecular Imaging and Translational Medicine, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Hehe Xiong
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory & Center for Molecular Imaging and Translational Medicine, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Heng Zhang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory & Center for Molecular Imaging and Translational Medicine, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Huaxiang Shen
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory & Center for Molecular Imaging and Translational Medicine, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Fantian Zeng
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory & Center for Molecular Imaging and Translational Medicine, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Changrong Shi
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory & Center for Molecular Imaging and Translational Medicine, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
| | - Zijian Zhou
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory & Center for Molecular Imaging and Translational Medicine, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China
| |
Collapse
|
32
|
Yang Y, Liang K, Zhou Z, Tu Y, Li M, Wang Z, Deng Y, Li J. Photoresponsive Bio-Heterojunctions Eliciting Immunogenicity to Prevent Infection Recurrence and Accelerating Chronic Wound Regeneration. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2410522. [PMID: 40035640 DOI: 10.1002/smll.202410522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 12/21/2024] [Indexed: 03/06/2025]
Abstract
Dynamic therapy utilizes reactive oxygen species (ROS) to antibacterial and enhance the innate immune system to treat bacterial infections. If ROS levels are too low, the elimination of pathogens and the enhancement of innate immunity cannot be achieved. However, excess accumulation of ROS may impact intracellular glutathione (GSH) levels, hindering T cell maturation and the establishment of immune memory. Herein, a multifunctional nanofiber membrane is designed, consisting of a polymer scaffold, MXene/CeO2 bio-heterojunctions (MX@Ce bio-HJs), and lactate oxidase (Lox) to balance the production of ROS, for the treatment of recurrent bacterial infections. In this system, MX@Ce bio-HJs upon near-infrared ray (NIR) generate photodynamic therapy, while Lox responds to the wound microenvironment exert chemodynamic therapy, synergistically produce ROS to rapidly eradicate bacteria, amplify the ability of dendritic cells to recognize and present antigens of bacterial fragments, and enhance innate immunity. Without NIR, MX@Ce bio-HJs showcase catalase-like and superoxide dismutase-like activities, scavenging subsequent ROS accumulation, promoting T cell maturation to form acquired immune memory, and combating recurrent bacterial infection. Such work highlights the potential to combat in situ bacterial infections and recurrent bacterial infections and inspires the development of future antibacterial therapies.
Collapse
Affiliation(s)
- Yingming Yang
- State Key Laboratory of Oral Diseases, School of Chemical Engineering, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, 610065, China
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Chengdu, 610041, China
| | - Kunneng Liang
- State Key Laboratory of Oral Diseases, School of Chemical Engineering, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, 610065, China
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Chengdu, 610041, China
| | - Zilin Zhou
- State Key Laboratory of Oral Diseases, School of Chemical Engineering, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, 610065, China
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Chengdu, 610041, China
| | - Yuanyuan Tu
- State Key Laboratory of Oral Diseases, School of Chemical Engineering, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, 610065, China
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Chengdu, 610041, China
| | - Meng Li
- State Key Laboratory of Oral Diseases, School of Chemical Engineering, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, 610065, China
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Chengdu, 610041, China
| | - Ziyou Wang
- State Key Laboratory of Oral Diseases, School of Chemical Engineering, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, 610065, China
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Chengdu, 610041, China
| | - Yi Deng
- State Key Laboratory of Oral Diseases, School of Chemical Engineering, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, 610065, China
- State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China
- Department of Mechanical Engineering, The University of Hong Kong, Hong Kong, 999077, China
| | - Jiyao Li
- State Key Laboratory of Oral Diseases, School of Chemical Engineering, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, 610065, China
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Chengdu, 610041, China
| |
Collapse
|
33
|
Jiang Y, Li S, Jiang L, Mu G, Jiang S. Immunomodulatory activity and molecular mechanisms of action of peptides derived from casein hydrolysate by alcalase and flavourzyme based on virtual screening. J Dairy Sci 2025; 108:2152-2168. [PMID: 39603497 DOI: 10.3168/jds.2024-25224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 11/04/2024] [Indexed: 11/29/2024]
Abstract
This study aimed to screen novel immunomodulatory peptides from casein hydrolysates (CH) using alcalase and flavorzyme by virtual screening, and their molecular mechanism were further studied. Based on the primary structural characteristics of immunomodulatory peptides, along with their hydrophobicity and isoelectric point, 3 novel immunomodulatory peptides (ALPMHIR, AMKPWIQPK, NPWDQVKR) were quickly found using virtual screening. These peptides exhibited strong interactions with TLR2/TLR4 through hydrogen bonding and hydrophobic interactions. Molecular docking verified that the key binding sites, such as Ile733, Ala732, and Phe774 in TLR2/TLR4 contributed to docking. Interestingly, the peptide AMKPWIQPK exhibited the strongest immunomodulatory activity and anti-inflammatory activity as 2-way immunomodulatory peptides. Based on western blot analysis and validation using specific inhibitors against MAPK/NF-κB signaling pathways, the results demonstrated that AMKPWIQPK could recognize the TLR2 and TLR4 receptor of the macrophages to upregulate the phospho-IκBα, phospho-p38, and phospho-p65, and further activated the MAPKs/NF-κB signaling pathways to enhance the immunomodulatory activity. These results confirmed that screening and optimizing immunomodulatory peptides by virtual screening and molecular docking were a novel and rapidly feasible method. The peptide AMKPWIQPK was expected to be used as natural-derived immunomodulatory active ingredients in nutritional health care and functional foods.
Collapse
Affiliation(s)
- Yutong Jiang
- School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China
| | - Siyi Li
- School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China
| | - Lai Jiang
- School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China
| | - Guangqing Mu
- School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China
| | - Shujuan Jiang
- School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China.
| |
Collapse
|
34
|
Xu Q, Jiang Y, Chen J, Wu J, Chen Y, Fan Q, Wang H, Yang Y, Pan J, Fang Q. Single Cell-Pair Proteomics for Decoding Immune-Cancer Cell Interactions. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2414769. [PMID: 39840604 PMCID: PMC11923901 DOI: 10.1002/advs.202414769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 01/02/2025] [Indexed: 01/23/2025]
Abstract
The efficacy of cancer immunotherapy is significantly influenced by the heterogeneity of individual tumors and immune responses. To investigate this phenomenon, a microfluidic platform is constructed for profiling immune-cancer cell interactions at the single-cell proteomics level for the first time. Based on the platform, a comprehensive workflow is proposed for achieving accurate single-cell pairing of an immune cell and a cancer cell with low cell damage and high success rate up to 95%, cell pair co-culture, and real-time microscopic monitoring of the cell-pair interactions, cell pair retrieval, mass spectrometry-based proteomic analysis of singe cell pairs, and decoupling of the proteomic information for each cell within the cell pair with the stable-isotope labeling method. With the workflow, the interactions of single natural killer (NK) cells and single K562 tumor cells are investigated based on real-time images and single cell-pair proteomics. Notably, an identification depth of over 1000 protein groups in a single cell-pair is achieved, leading to the discovery of sub-clusters of NK cells with different functions and the identification of important biomarkers for cancer treatments. This demonstrates the unique capability of the present platform in providing substantial and comprehensive datasets for profiling immune-cancer cell interactions, discovering heterogeneous immune responses, and predicting biomarkers in the study of cancer immunotherapy.
Collapse
Affiliation(s)
- Qin‐Qin Xu
- Institute of Microanalytical SystemsDepartment of ChemistryZhejiang UniversityHangzhou310058China
| | - Yi‐Rong Jiang
- Institute of Microanalytical SystemsDepartment of ChemistryZhejiang UniversityHangzhou310058China
| | - Jian‐Bo Chen
- Institute of Microanalytical SystemsDepartment of ChemistryZhejiang UniversityHangzhou310058China
| | - Jie Wu
- Institute of Microanalytical SystemsDepartment of ChemistryZhejiang UniversityHangzhou310058China
| | - Yi‐Xue Chen
- Institute of Microanalytical SystemsDepartment of ChemistryZhejiang UniversityHangzhou310058China
| | - Qian‐Xi Fan
- Institute of Microanalytical SystemsDepartment of ChemistryZhejiang UniversityHangzhou310058China
| | - Hui‐Feng Wang
- Institute of Microanalytical SystemsDepartment of ChemistryZhejiang UniversityHangzhou310058China
- Key Laboratory of Excited‐State Materials of Zhejiang ProvinceZhejiang UniversityHangzhou310007China
| | - Yi Yang
- Single‐cell Proteomics Research CenterZJU‐Hangzhou Global Scientific and Technological Innovation CenterHangzhou311200China
- Engineering Research Center of Functional Materials Intelligent Manufacturing of Zhejiang ProvinceHangzhou311200China
| | - Jian‐Zhang Pan
- Institute of Microanalytical SystemsDepartment of ChemistryZhejiang UniversityHangzhou310058China
- Single‐cell Proteomics Research CenterZJU‐Hangzhou Global Scientific and Technological Innovation CenterHangzhou311200China
- Engineering Research Center of Functional Materials Intelligent Manufacturing of Zhejiang ProvinceHangzhou311200China
| | - Qun Fang
- Institute of Microanalytical SystemsDepartment of ChemistryZhejiang UniversityHangzhou310058China
- Key Laboratory of Excited‐State Materials of Zhejiang ProvinceZhejiang UniversityHangzhou310007China
- Single‐cell Proteomics Research CenterZJU‐Hangzhou Global Scientific and Technological Innovation CenterHangzhou311200China
- Engineering Research Center of Functional Materials Intelligent Manufacturing of Zhejiang ProvinceHangzhou311200China
- Key Laboratory for Biomedical Engineering of Ministry of Education, Cancer CenterZhejiang UniversityHangzhou310007China
| |
Collapse
|
35
|
Dash UC, Nayak V, Navani HS, Samal RR, Agrawal P, Singh AK, Majhi S, Mogare DG, Duttaroy AK, Jena AB. Understanding the molecular bridges between the drugs and immune cell. Pharmacol Ther 2025; 267:108805. [PMID: 39908660 DOI: 10.1016/j.pharmthera.2025.108805] [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: 09/09/2024] [Revised: 01/11/2025] [Accepted: 01/21/2025] [Indexed: 02/07/2025]
Abstract
The interactions of drugs with the host's immune cells determine the drug's efficacy and adverse effects in patients. Nonsteroidal Anti-Inflammatory Drugs (NSAID), such as corticosteroids, NSAIDs, and immunosuppressants, affect the immune cells and alter the immune response. Molecularly, drugs can interact with immune cells via cell surface receptors, changing the antigen presentation by modifying the co-stimulatory molecules and interacting with the signaling pathways of T cells, B cells, Natural killer (NK) cells, mast cells, basophils, and macrophages. Immunotoxicity, resulting from drug-induced changes in redox status, generation of Reactive Oxygen Species (ROS)/Reactive Nitrogen Species (RNS), and alterations in antioxidant enzymes within immune cells, leads to immunodeficiency. This, in turn, causes allergic reactions, autoimmune diseases, and cytokine release syndrome (CRS). The treatment options should include the evaluation of immune status and utilization of the concept of pharmacogenomics to minimize the chances of immunotoxicity. Many strategies in redox, like targeting the redox pathway or using redox-active agents, are available for the modulation of the immune system and developing drugs. Case studies highlight significant drug-immune cell interactions and patient outcomes, underscoring the importance of understanding these complexities. The future direction focuses on the drugs to deliver antiviral therapy, new approaches to immunomodulation, and modern technologies for increasing antidote effects with reduced toxicity. In conclusion, in-depth knowledge of the interaction between drugs and immune cells is critical to protect the patient from the adverse effects of the drug and improve therapeutic outcomes of the treatment process. This review focuses on the multifaceted interactions of drugs and their consequences at the cellular levels of immune cells.
Collapse
Affiliation(s)
- Umesh Chandra Dash
- School of Biotechnology, Campus 11, Kalinga Institute of Industrial Technology (KIIT) Deemed to be University, Bhubaneswar, Odisha 751024, India
| | - Vinayak Nayak
- Department of Biotechnology, Indian Institute of Technology Hyderabad, Kandi, Sangareddy 502284, India
| | - Hiten Shanker Navani
- Biological Materials Laboratory, CSIR- Central Leather Research Institute, Adyar, Chennai 600020, India
| | - Rashmi Rekha Samal
- CSIR-Institute of Minerals & Materials Technology, Bhubaneswar 751 013, India
| | - Palak Agrawal
- Unit de Microbiologie Structurale, Institut Pasteur, Paris, France
| | - Anup Kumar Singh
- National Centre for Cell Science, Savitribai Phule Pune University Campus, Ganeshkhind, Pune, India
| | - Sanatan Majhi
- Post Graduate Department of Biotechnology, Utkal University, Bhubaneswar 751004, Odisha, India
| | - Devraj Ganpat Mogare
- National Centre for Cell Science, Savitribai Phule Pune University Campus, Ganeshkhind, Pune, India
| | - Asim K Duttaroy
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, 0317 Oslo, Norway.
| | - Atala Bihari Jena
- National Centre for Cell Science, Savitribai Phule Pune University Campus, Ganeshkhind, Pune, India
| |
Collapse
|
36
|
Chen Z, Fan J, Chen X, Yang K, Wang K. Oxidative Stress and Redox Signaling in Gastric Cancer: From Mechanisms to Therapeutic Implications. Antioxidants (Basel) 2025; 14:258. [PMID: 40227215 PMCID: PMC11939249 DOI: 10.3390/antiox14030258] [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: 01/27/2025] [Revised: 02/18/2025] [Accepted: 02/22/2025] [Indexed: 04/15/2025] Open
Abstract
Oxidative stress, which is characterized by an imbalance between reactive oxygen species (ROS) production and antioxidant defenses, has critical roles in the initiation, progression, and treatment of gastric cancer. On the one hand, an excessive ROS accumulation induces oxidative damage and cancer cell death. On the other hand, moderate levels of ROS cause genetic mutations and dysregulation of signaling pathways to promote proliferation, inflammation, angiogenesis, and metastasis in gastric cancer. Notably, emerging evidence has revealed that ROS also mediate oxidative post-translational modifications (oxPTMs) of redox-sensitive proteins, which can directly affect protein functions and regulate redox signaling in cancer cells. Therefore, elucidating the regulatory mechanisms of oxidative stress and redox signaling in gastric cancer holds great promise to identify novel therapeutic targets or redox-targeting strategies. This review will summarize the mechanisms of oxidative stress in regulating the hallmarks of gastric cancer and highlight the roles of ROS-mediated oxPTMs in gastric cancer. In addition, we will discuss emerging strategies targeting oxidative stress for the treatment of gastric cancer, with an emphasis on the use of bioactive natural products and nanomaterials.
Collapse
Affiliation(s)
- Zehua Chen
- Department of General Surgery and Laboratory of Gastric Cancer, West China School of Basic Medical Sciences & Forensic Medicine, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China; (Z.C.); (J.F.); (X.C.)
- Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jiawu Fan
- Department of General Surgery and Laboratory of Gastric Cancer, West China School of Basic Medical Sciences & Forensic Medicine, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China; (Z.C.); (J.F.); (X.C.)
| | - Xiaolong Chen
- Department of General Surgery and Laboratory of Gastric Cancer, West China School of Basic Medical Sciences & Forensic Medicine, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China; (Z.C.); (J.F.); (X.C.)
- Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Kun Yang
- Department of General Surgery and Laboratory of Gastric Cancer, West China School of Basic Medical Sciences & Forensic Medicine, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China; (Z.C.); (J.F.); (X.C.)
- Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Kui Wang
- Department of General Surgery and Laboratory of Gastric Cancer, West China School of Basic Medical Sciences & Forensic Medicine, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China; (Z.C.); (J.F.); (X.C.)
| |
Collapse
|
37
|
Wang H, Chen B, Xiao P, Han D, Gao B, Yan Y, Zhao R, Pan T, Zhang J, Zhou M, Lv L, Gao H. Yersiniabactin produced by Escherichia coli promotes intestinal inflammation through lipid peroxidation and ferroptosis. Front Microbiol 2025; 16:1542801. [PMID: 40034497 PMCID: PMC11872927 DOI: 10.3389/fmicb.2025.1542801] [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: 12/10/2024] [Accepted: 02/03/2025] [Indexed: 03/05/2025] Open
Abstract
Escherichia coli (E. coli), a major foodborne pathogen, poses significant risks to public health by causing gastrointestinal diseases. Among its virulence factors, Yersiniabactin (Ybt), a siderophore, plays a crucial role in iron acquisition and enhancing intestinal colonization. Despite previous studies highlighting E. coli-Ybt's involvement in inflammation, its exact mechanisms remain unclear. This study investigates how Ybt contributes to intestinal inflammation through ferroptosis, using both in vitro and in vivo models. Our findings demonstrate that Ybt promotes oxidative stress, lipid peroxidation, inflammation, and iron accumulation in intestinal epithelial cells, leading to ferroptosis. Mechanistically, Ybt suppresses the Keap1/Nrf2 pathway, amplifying reactive oxygen species (ROS) and activating the TNF/NF-κB pathway, which drives inflammation. Moreover, Ybt induces lipid peroxidation via the arachidonic acid pathway, producing 6-trans-leukotriene B4 (6-transLTB4), which exacerbates inflammation and ferroptosis. Exogenous 6-transLTB4 further intensifies this cascade. Additionally, Ybt disrupts iron efflux by suppressing FPN1 expression, causing excessive intracellular iron accumulation. Using tree shrews as an in vivo model, we confirm that Ybt-induced ferroptosis significantly aggravates intestinal inflammation. These findings underscore the pathogenic role of Ybt in E. coli-induced intestinal injury and highlight ferroptosis as a novel mechanism contributing to gut health disruption. This study provides new insights into the molecular pathways of E. coli infection, with implications for therapeutic strategies targeting ferroptosis in intestinal diseases.
Collapse
Affiliation(s)
- Hao Wang
- College of Food Science and Technology, Yunnan Agricultural University, Kunming, China
| | - Bingxun Chen
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming, China
| | - Peng Xiao
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming, China
| | - Dongmei Han
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming, China
| | - Bin Gao
- College of Food Science and Technology, Yunnan Agricultural University, Kunming, China
| | - Yulin Yan
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming, China
| | - Ru Zhao
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming, China
| | - Tianling Pan
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming, China
| | - Jingsong Zhang
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming, China
| | - Meng Zhou
- College of Food Science and Technology, Yunnan Agricultural University, Kunming, China
| | - Longbao Lv
- National Resource Center for Non-Human Primates, National Research Facility for Phenotypic and Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Hong Gao
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming, China
| |
Collapse
|
38
|
Lee C, Khan R, Mantsounga CS, Sharma S, Pierce J, Amelotte E, Butler CA, Farinha A, Parry C, Caballero O, Morrison JA, Uppuluri S, Whyte JJ, Kennedy JL, Zhang X, Choudhary G, Olson RM, Morrison AR. IL-1β-driven NF-κB transcription of ACE2 as a Mechanism of Macrophage Infection by SARS-CoV-2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.12.24.630260. [PMID: 39763770 PMCID: PMC11703209 DOI: 10.1101/2024.12.24.630260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
Coronavirus disease 2019 (COVID-19), caused by infection with the enveloped RNA betacoronavirus, SARS-CoV-2, led to a global pandemic involving over 7 million deaths. Macrophage inflammatory responses impact COVID-19 severity; however, it is unclear whether macrophages are infected by SARS-CoV-2. We sought to identify mechanisms regulating macrophage expression of ACE2, the primary receptor for SARS-CoV-2, and to determine if macrophages are susceptible to productive infection. We developed a humanized ACE2 (hACE2) mouse whereby hACE2 cDNA was cloned into the mouse ACE2 locus under control of the native promoter. We validated the susceptibility of hACE2 mice to SARS-CoV-2 infection relative to wild-type mice and an established K18-hACE2 model of acute fulminating disease. Intranasal exposure to SARS-CoV-2 led to pulmonary consolidations with cellular infiltrate, edema, and hemorrhage, consistent with pneumonia, yet unlike the K18-hACE2 model, hACE2 mice survived and maintained stable weight. Infected hACE2 mice also exhibited a unique plasma chemokine, cytokine, and growth factor inflammatory signature relative to K18-hACE2 mice. Infected hACE2 mice demonstrated evidence of viral replication in infiltrating lung macrophages, and infection of macrophages in vitro revealed a transcriptional profile indicative of altered RNA and ribosomal processing machinery as well as activated cellular antiviral defense. Macrophage IL-1β-driven NF-κB transcription of ACE2 was an important mechanism of dynamic ACE2 upregulation, promoting macrophage susceptibility to infection. Experimental models of COVID-19 that make use of native hACE2 expression will allow for mechanistic insight into factors that can either promote host resilience or increase susceptibility to worsening severity of infection.
Collapse
Affiliation(s)
- Cadence Lee
- Vascular Research Laboratory, Providence VA Medical Center, Providence, Rhode Island 02908, USA
- Ocean State Research Institute, Inc., Providence, Rhode Island 02908, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, Rhode Island 02903, USA
| | - Rachel Khan
- Vascular Research Laboratory, Providence VA Medical Center, Providence, Rhode Island 02908, USA
- Ocean State Research Institute, Inc., Providence, Rhode Island 02908, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, Rhode Island 02903, USA
| | - Chris S. Mantsounga
- Vascular Research Laboratory, Providence VA Medical Center, Providence, Rhode Island 02908, USA
- Ocean State Research Institute, Inc., Providence, Rhode Island 02908, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, Rhode Island 02903, USA
| | - Sheila Sharma
- Vascular Research Laboratory, Providence VA Medical Center, Providence, Rhode Island 02908, USA
- Ocean State Research Institute, Inc., Providence, Rhode Island 02908, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, Rhode Island 02903, USA
| | - Julia Pierce
- Vascular Research Laboratory, Providence VA Medical Center, Providence, Rhode Island 02908, USA
- Ocean State Research Institute, Inc., Providence, Rhode Island 02908, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, Rhode Island 02903, USA
| | - Elizabeth Amelotte
- Vascular Research Laboratory, Providence VA Medical Center, Providence, Rhode Island 02908, USA
- Ocean State Research Institute, Inc., Providence, Rhode Island 02908, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, Rhode Island 02903, USA
| | - Celia A. Butler
- Vascular Research Laboratory, Providence VA Medical Center, Providence, Rhode Island 02908, USA
- Ocean State Research Institute, Inc., Providence, Rhode Island 02908, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, Rhode Island 02903, USA
| | - Andrew Farinha
- Vascular Research Laboratory, Providence VA Medical Center, Providence, Rhode Island 02908, USA
- Ocean State Research Institute, Inc., Providence, Rhode Island 02908, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, Rhode Island 02903, USA
| | - Crystal Parry
- Vascular Research Laboratory, Providence VA Medical Center, Providence, Rhode Island 02908, USA
- Ocean State Research Institute, Inc., Providence, Rhode Island 02908, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, Rhode Island 02903, USA
| | - Olivya Caballero
- Vascular Research Laboratory, Providence VA Medical Center, Providence, Rhode Island 02908, USA
- Ocean State Research Institute, Inc., Providence, Rhode Island 02908, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, Rhode Island 02903, USA
| | - Jeremi A. Morrison
- Vascular Research Laboratory, Providence VA Medical Center, Providence, Rhode Island 02908, USA
- Ocean State Research Institute, Inc., Providence, Rhode Island 02908, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, Rhode Island 02903, USA
| | - Saketh Uppuluri
- Vascular Research Laboratory, Providence VA Medical Center, Providence, Rhode Island 02908, USA
- Ocean State Research Institute, Inc., Providence, Rhode Island 02908, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, Rhode Island 02903, USA
| | - Jeffrey J. Whyte
- Department of Veterinary Pathobiology, University of Missouri College of Veterinary Medicine, Columbia, Missouri, USA
- Laboratory for Infectious Disease Research, University of Missouri Division of Research, Innovation and Impact, Columbia, Missouri, USA
| | - Joshua L. Kennedy
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
- Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
- Arkansas Children’s Research Institute, Little Rock, Arkansas, USA
| | - Xuming Zhang
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Gaurav Choudhary
- Vascular Research Laboratory, Providence VA Medical Center, Providence, Rhode Island 02908, USA
- Ocean State Research Institute, Inc., Providence, Rhode Island 02908, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, Rhode Island 02903, USA
- Cardiovascular Research Center, Lifespan Cardiovascular Research Institute, Rhode Island Hospital, Providence, Rhode Island, USA
| | - Rachel M. Olson
- Department of Veterinary Pathobiology, University of Missouri College of Veterinary Medicine, Columbia, Missouri, USA
- Laboratory for Infectious Disease Research, University of Missouri Division of Research, Innovation and Impact, Columbia, Missouri, USA
| | - Alan R. Morrison
- Vascular Research Laboratory, Providence VA Medical Center, Providence, Rhode Island 02908, USA
- Ocean State Research Institute, Inc., Providence, Rhode Island 02908, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, Rhode Island 02903, USA
- Lead contact and corresponding author
| |
Collapse
|
39
|
Jin Y, Liu H, Wang Y, Zhang R, Wang Q, Wang Y, Cui H, Wang X, Bian Y. Pathogenesis and treatment of colitis-associated colorectal cancer: Insights from Traditional Chinese Medicine. JOURNAL OF ETHNOPHARMACOLOGY 2025; 338:119096. [PMID: 39532222 DOI: 10.1016/j.jep.2024.119096] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 10/11/2024] [Accepted: 11/09/2024] [Indexed: 11/16/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Inflammatory Bowel Disease (IBD) is an inflammatory intestinal disease, and with prolonged illness duration, the annual risk of IBD progressing to colitis-associated colorectal cancer (CAC) gradually increases. In recent years, there has been a noticeable trend towards the application of traditional Chinese medicine (TCM) in the treatment of CAC. AIM OF THIS REVIEW This comprehensive review summarizes the pathogenesis of CAC and details the therapeutic benefits of TCM in treating CAC, including various TCM prescriptions and ingredients, establishing the theoretical foundation for the application of TCM in CAC treatment. METHODS We assessed literature published before March 24, 2024, from several databases, including Web of Science, PubMed, Scopus and Google Scholar. The keywords used include "traditional Chinese medicine", "traditional Chinese medicine prescriptions", "traditional Chinese medicine ingredients", "herbal medicine", "colitis-associated colorectal cancer", "inflammatory bowel disease", "colorectal cancer" and "colitis-cancer transformation". We conducted a comprehensive collection and collation of pertinent scientific articles from various databases, focusing on the efficacy of TCM in the prevention and treatment of "colitis-cancer transformation". RESULTS This paper provides a concise summary and thorough analysis of twenty-eight prescriptions and ingredients of TCM for the prevention and treatment of CAC, based on existing experimental and clinical research. There are positive signs that TCM can effectively prevent and treat the "colitis-cancer transformation" through repairing the intestinal mucosal barrier, correcting intestinal flora imbalance, and regulating intestinal immune responses. CONCLUSION TCM possesses comprehensive regulatory advantages that are multifaceted, multilevel, and multitarget. It has a definite curative effect in the prevention and treatment of CAC. It is essential to enhance the clinical efficacy of TCM in the prevention and treatment of CAC based on syndrome differentiation and treatment, with the assistance of modern medicine.
Collapse
Affiliation(s)
- Yutong Jin
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Haizhao Liu
- Department of Integrated Traditional Chinese and Western Medicine, Tianjin First Central Hospital, Nankai University, Tianjin, 300192, China
| | - Yuhui Wang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Ruixuan Zhang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Qiaochu Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300073, China
| | - Yao Wang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; School of Basic Medical Sciences, Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - Huantian Cui
- First School of Clinical Medicine, Yunnan University of Chinese Medicine, Kunming, 650500, China.
| | - Xiangling Wang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| | - Yuhong Bian
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| |
Collapse
|
40
|
Chen Z, Zhou Y, Li L, Ma W, Li Y, Yang Z. Activatable Molecular Probes With Clinical Promise for NIR-II Fluorescent Imaging. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2411787. [PMID: 39707663 DOI: 10.1002/smll.202411787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 12/09/2024] [Indexed: 12/23/2024]
Abstract
The second near-infrared window (NIR-II) fluorescence imaging has been widely adopted in basic scientific research and preclinical applications due to its exceptional spatiotemporal resolution and deep tissue penetration. Among the various fluorescent agents, organic small-molecule fluorophores are considered the most promising candidates for clinical translation, owing to their well-defined chemical structures, tunable optical properties, and excellent biocompatibility. However, many currently available NIR-II fluorophores exhibit an "always-on" fluorescence signal, which leads to background noise and compromises diagnostic accuracy during disease detection. Developing NIR-II activatable organic small-molecule fluorescent probes (AOSFPs) for accurately reporting pathological changes is key to advancing NIR-II fluorescence imaging toward clinical application. This review summarizes the rational design strategies for NIR-II AOSFPs based on four core structures (cyanine, hemicyanine, xanthene, and BODIPY). These NIR-II AOSFPs hold substantial potential for clinical translation. Furthermore, the recent advances in NIR-II AOSFPs for NIR-II bioimaging are comprehensively reviewed, offering clear guidance and direction for their further development. Finally, the prospective efforts to advance NIR-II AOSFPs for clinical applications are outlined.
Collapse
Affiliation(s)
- Zikang Chen
- Department of Pharmacy, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518000, China
| | - Yongjie Zhou
- Department of Interventional Radiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Li Li
- Department of Pharmacy, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518000, China
| | - Wen Ma
- Strait Institute of Flexible Electronics (SIFE Future Technologies), Fujian Key Laboratory of Flexible Electronics, Fujian Normal University and Strait Laboratory of Flexible Electronics (SLoFE), Fuzhou, 350117, China
| | - Yuzhen Li
- Department of Pharmacy, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518000, China
| | - Zhen Yang
- Strait Institute of Flexible Electronics (SIFE Future Technologies), Fujian Key Laboratory of Flexible Electronics, Fujian Normal University and Strait Laboratory of Flexible Electronics (SLoFE), Fuzhou, 350117, China
| |
Collapse
|
41
|
Wu J, Hu Y, Zhao N, Yang W, Chen Z. The active roles of Rhodotorula mucilaginosa ZTHY2 in regulating antioxidant capacity and immune function of Leizhou black ducks. Front Vet Sci 2025; 12:1494892. [PMID: 39950088 PMCID: PMC11821949 DOI: 10.3389/fvets.2025.1494892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 01/15/2025] [Indexed: 02/16/2025] Open
Abstract
Previous studies in mice have demonstrated that Rhodotorula mucilaginosa ZTHY2 can promote animal growth, enhance antioxidant and immune functions, and regulate intestinal flora in our laboratory. This study focuses on the Leizhou black duck, a local breed in Zhanjiang, to evaluate the effects of Rhodotorula mucilaginosa ZTHY2 on its growth, antioxidant capacity, and immune function. A total of 150 1-day-old male Leizhou black ducks, of similar size and healthy, were selected for this study and randomly assigned to five treatment groups. Each group contained three replicates with ten birds each. The control group (Control) was given a standard basal diet, while the RM group received a diet supplemented with ZTHY2 at concentrations of 2 × 107 (RM1), 2 × 108(RM2), or 2 × 109(RM3) CFU/kg, respectively. The LA group was supplemented with 2 × 109 CFU/kg of Lactobacillus acidophilus in addition to the basal diet. The feeding trial lasted 42 days. The analysis revealed significant improvements in the average body weight for the RM2 and RM3 groups, which were significantly higher than that of the control group (p < 0.05 and p < 0.01). Treatment with ZTHY2 induced a dose-dependent elevation in superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GSH-Px), and SOD activities, and a reduction in malondialdehyde (MDA) content in the serum at 42 days. The serum levels of complement components C3 and C4, immunoglobulin IgG, and cytokines IFN-γ, IL-2, IL-4, IL-6, and TNF-α were significantly increased in Leizhou black ducks treated with ZTHY2 at 42-days post-treatment, with the therapeutic effect becoming more pronounced as the duration of the experiment prolonged. The greatest impact was observed at a dosage of 2 × 109 CFU/kg of ZTHY2. Moreover, ZTHY2 modulated the mRNA expression profiles of these cytokines in the thymus, spleen, and bursa, thereby sustaining the balance of immune dynamics. In summary, the supplementation of Rhodotorula mucilaginosa ZTHY2 at a dosage of 2 × 109 CFU/kg had been found to most effectively enhance the growth performance of Leizhou black ducks by optimizing their immune function and antioxidant capacity.
Collapse
Affiliation(s)
- Jiang Wu
- Department of Veterinary Medicine, College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang, China
| | - Yingxin Hu
- Department of Veterinary Medicine, College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang, China
| | - Namula Zhao
- Department of Veterinary Medicine, College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang, China
| | - Wei Yang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Zhibao Chen
- Department of Veterinary Medicine, College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang, China
- South China Branch of National Saline-Alkali Tolerant Rice Technology Innovation Center Zhanjiang, Zhanjiang, China
| |
Collapse
|
42
|
Chandimali N, Bak SG, Park EH, Lim HJ, Won YS, Kim EK, Park SI, Lee SJ. Free radicals and their impact on health and antioxidant defenses: a review. Cell Death Discov 2025; 11:19. [PMID: 39856066 PMCID: PMC11760946 DOI: 10.1038/s41420-024-02278-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 12/10/2024] [Accepted: 12/17/2024] [Indexed: 01/27/2025] Open
Abstract
Free radicals, characterized by the presence of unpaired electrons, are highly reactive species that play a significant role in human health. These molecules can be generated through various endogenous processes, such as mitochondrial respiration and immune cell activation, as well as exogenous sources, including radiation, pollution, and smoking. While free radicals are essential for certain physiological processes, such as cell signaling and immune defense, their overproduction can disrupt the delicate balance between oxidants and antioxidants, leading to oxidative stress. Oxidative stress results in the damage of critical biomolecules like DNA, proteins, and lipids, contributing to the pathogenesis of various diseases. Chronic conditions such as cancer, cardiovascular diseases, neurodegenerative disorders, and inflammatory diseases have been strongly associated with the harmful effects of free radicals. This review provides a comprehensive overview of the characteristics and types of free radicals, their mechanisms of formation, and biological impacts. Additionally, we explore natural compounds and extracts studied for their antioxidant properties, offering potential therapeutic avenues for managing free radical-induced damage. Future research directions are also discussed to advance our understanding and treatment of free radical-associated diseases.
Collapse
Affiliation(s)
- Nisansala Chandimali
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Jeongeup, 56212, Korea
- Applied Biological Engineering, KRIBB School of Biotechnology, University of Science and Technology, Daejeon, 34113, Korea
| | - Seon Gyeong Bak
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Jeongeup, 56212, Korea
| | - Eun Hyun Park
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Jeongeup, 56212, Korea
- Department of Veterinary Pathology, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju, 61186, Korea
| | - Hyung-Jin Lim
- Scripps Korea Antibody Institute, Chuncheon, 24341, Korea
| | - Yeong-Seon Won
- Division of Research Management, Department of Bioresource Industrialization, Honam National Institute of Biological Resource, Mokpo, 58762, Korea
| | - Eun-Kyung Kim
- Nutritional Education Major, Graduate School of Education, Dong-A University, Busan, 49315, Korea
| | - Sang-Ik Park
- Department of Veterinary Pathology, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju, 61186, Korea.
| | - Seung Jae Lee
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Jeongeup, 56212, Korea.
- Applied Biological Engineering, KRIBB School of Biotechnology, University of Science and Technology, Daejeon, 34113, Korea.
| |
Collapse
|
43
|
Aghila Rani KG, Soares NC, Rahman B, Giddey AD, Al-Hroub HM, Semreen MH, Al Kawas S. Medwakh smoking induces alterations in salivary proteins and cytokine expression: a clinical exploratory proteomics investigation. Clin Proteomics 2025; 22:2. [PMID: 39819313 PMCID: PMC11740365 DOI: 10.1186/s12014-024-09520-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Accepted: 12/16/2024] [Indexed: 01/19/2025] Open
Abstract
BACKGROUND Medwakh smoking has radically expanded among youth in the Middle East and around the world. The rising popularity of medwakh/dokha usage is linked to the onset of several chronic illnesses including cardiovascular diseases and cancers. Medwakh smoking is reported to increase the risk of inflammation in the lower respiratory tract owing to oxidative burden. To date, there are no reported studies investigating the impact of medwakh smoking on salivary protein profile. The current study aims to elucidate alterations in the salivary proteome profile of medwakh smokers. METHODS Saliva samples collected from 33 medwakh smokers and 30 non-smokers were subjected to proteomic analysis using UHPLC-ESI-QTOF-MS. Saliva samples were further subjected to validatory experiments involving analysis of inflammatory cytokine profile using LEGENDplex™ Human Essential Immune Response Panel. RESULTS Statistical analysis revealed alterations in the abundance of 74 key proteins including immune mediators and inflammatory markers in medwakh smokers (Accession: PXD045901). Proteins involved in building oxidative stress, alterations in cell anchorage, and cell metabolic processes were enhanced in medwakh smokers. Salivary immune response evaluation further validated the proteome findings, revealing significantly higher levels of IL-1β, IL-12p70, IL-23, IFN-γ (Th1 cytokines), IL-6 (Th2 cytokine), and MCP-1 (chemokine) in medwakh smokers. In addition, a substantial increase in abundance of involucrin suggesting a plausible stratified squamous cell differentiation and increased cell lysis in the oral cavity of medwakh smokers akin to chronic obstructive pulmonary diseases (COPD). The protein-metabolite joint pathway analysis further showed significantly enriched differentially expressed proteins and metabolites of glycolysis/gluconeogenesis, pentose phosphate, fructose and mannose, nicotinate and nicotinamide, and glutathione metabolism pathways among medwakh smokers. CONCLUSIONS The findings of the study provide valuable insights on potential perturbations in various key immune molecules, cytokines, and signaling pathways among medwakh smokers. Medwakh smokers displayed elevated inflammation, increased oxidative stress and defective antioxidant responses, dysregulated energy metabolism, and alterations in proteins related to cell adhesion, migration, differentiation, and proliferation. The findings of study underscore the urgent need for comprehensive public health interventions among youth by raising awareness, implementing effective smoking cessation programs, and promoting healthy lifestyle to safeguard the well-being of individuals and communities worldwide.
Collapse
Affiliation(s)
- K G Aghila Rani
- Research Institute for Medical and Health Sciences, University of Sharjah, P.O. Box 27272, Sharjah, UAE
| | - Nelson C Soares
- Center for Applied and Translational Genomics (CATG), Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai Health, Dubai, UAE
- College of Medine, Mohammed bin Rashid Al Maktoum University of Medicine and Health Sciences, Dubai Health, Dubai, UAE
- Department of Human Genetics, National Institute of Health Doutor Ricardo Jorge (INSA), Lisbon, Portugal
| | - Betul Rahman
- Research Institute for Medical and Health Sciences, University of Sharjah, P.O. Box 27272, Sharjah, UAE
- Department of Preventive and Restorative Dentistry, College of Dental Medicine, University of Sharjah, Sharjah, UAE
| | - Alexander D Giddey
- Center for Applied and Translational Genomics (CATG), Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai Health, Dubai, UAE
| | - Hamza M Al-Hroub
- Research Institute for Medical and Health Sciences, University of Sharjah, P.O. Box 27272, Sharjah, UAE
| | - Mohammad H Semreen
- Research Institute for Medical and Health Sciences, University of Sharjah, P.O. Box 27272, Sharjah, UAE.
| | - Sausan Al Kawas
- Research Institute for Medical and Health Sciences, University of Sharjah, P.O. Box 27272, Sharjah, UAE.
- Department of Oral and Craniofacial Health Sciences, College of Dental Medicine, University of Sharjah, Sharjah, UAE.
| |
Collapse
|
44
|
Gu C, Yao T, Dong C, Chen Z, Wei W, Li X, Niu Q, Yan Y, Hu Y. Inflammation mediates the adverse effects of urinary phthalate exposure on metabolic disease risk: Results from NHANES 2005-2016. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 290:117577. [PMID: 39729938 DOI: 10.1016/j.ecoenv.2024.117577] [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: 10/06/2024] [Revised: 11/29/2024] [Accepted: 12/17/2024] [Indexed: 12/29/2024]
Abstract
BACKGROUND Phthalates are a category of chemicals commonly utilized in various industrial applications and everyday products. Their associations with health issues remains a significant concern. Although some studies have suggested associations between phthalates and metabolic diseases, the current understanding of the associations is still limited, especially the lack of effects of mixed exposure. METHODS This cross-sectional study included information from 9217 participants in National Health and Nutrition Examination Survey (NHANES) from 2005 to 2016. Multivariate logistic regression was used to explore the associations between single phthalate exposure and obesity and its complications. Weighted quantile sum (WQS) regression and Quantile G-Computation (Qgcomp) models were used to further analyze the associations between mixed phthalate exposure and obesity and its complications. Mediated analysis was used to explore the mediating role of immune cells in the relationship between phthalate exposure and obesity and its complications. RESULTS MiBP, MCOP and MBzP were associated with an increased risk of obesity. MiBP and MCOP were associated with an increased risk of abdominal obesity. MCNP, MCOP, MEHHP, MEOHP and MECPP were positively associated with T2DM. Mixed phthalate exposure was positively associated with obesity and T2DM. Monocytes mediated the effects of MiBP, MEHP and MBzP on obesity, explaining 7.94 %, -2.32 % and 6.69% of the total effect, respectively. CONCLUSIONS This study revealed a significant association between mixed phthalate exposure and obesity and its complications, underlining the importance of considering the interactions of these compounds. The synergistic effects of multiple phthalates may exacerbate health risks.
Collapse
Affiliation(s)
- Chenyang Gu
- Department of Preventive Medicine, School of Medicine, Shihezi University, Shihezi, Xinjiang, China
| | - Teng Yao
- Department of Preventive Medicine, School of Medicine, Shihezi University, Shihezi, Xinjiang, China; Key Laboratory for Prevention and Control of Emerging Infectious Diseases and Public Health Security, the Xinjiang Production and Construction Corps, China; Key Laboratory of Preventive Medicine, Shihezi University, Shihezi, Xinjiang, China
| | - Chenxian Dong
- Department of Preventive Medicine, School of Medicine, Shihezi University, Shihezi, Xinjiang, China; Key Laboratory for Prevention and Control of Emerging Infectious Diseases and Public Health Security, the Xinjiang Production and Construction Corps, China; Key Laboratory of Preventive Medicine, Shihezi University, Shihezi, Xinjiang, China
| | - Zuhai Chen
- Department of Preventive Medicine, School of Medicine, Shihezi University, Shihezi, Xinjiang, China
| | - Wanting Wei
- Department of Preventive Medicine, School of Medicine, Shihezi University, Shihezi, Xinjiang, China
| | - Xiaoju Li
- Department of Preventive Medicine, School of Medicine, Shihezi University, Shihezi, Xinjiang, China; Key Laboratory for Prevention and Control of Emerging Infectious Diseases and Public Health Security, the Xinjiang Production and Construction Corps, China; Key Laboratory of Preventive Medicine, Shihezi University, Shihezi, Xinjiang, China; Key Laboratory of Xinjiang Endemic and Ethnic Diseases (Ministry of Education), School of Medicine, Shihezi University, Shihezi, Xinjiang, China
| | - Qiang Niu
- Department of Preventive Medicine, School of Medicine, Shihezi University, Shihezi, Xinjiang, China; Key Laboratory for Prevention and Control of Emerging Infectious Diseases and Public Health Security, the Xinjiang Production and Construction Corps, China; Key Laboratory of Preventive Medicine, Shihezi University, Shihezi, Xinjiang, China; Key Laboratory of Xinjiang Endemic and Ethnic Diseases (Ministry of Education), School of Medicine, Shihezi University, Shihezi, Xinjiang, China.
| | - Yizhong Yan
- Department of Preventive Medicine, School of Medicine, Shihezi University, Shihezi, Xinjiang, China; Key Laboratory for Prevention and Control of Emerging Infectious Diseases and Public Health Security, the Xinjiang Production and Construction Corps, China; Key Laboratory of Preventive Medicine, Shihezi University, Shihezi, Xinjiang, China; Key Laboratory of Xinjiang Endemic and Ethnic Diseases (Ministry of Education), School of Medicine, Shihezi University, Shihezi, Xinjiang, China.
| | - Yunhua Hu
- Department of Preventive Medicine, School of Medicine, Shihezi University, Shihezi, Xinjiang, China; Key Laboratory for Prevention and Control of Emerging Infectious Diseases and Public Health Security, the Xinjiang Production and Construction Corps, China; Key Laboratory of Preventive Medicine, Shihezi University, Shihezi, Xinjiang, China; Key Laboratory of Xinjiang Endemic and Ethnic Diseases (Ministry of Education), School of Medicine, Shihezi University, Shihezi, Xinjiang, China.
| |
Collapse
|
45
|
Kim H, Kim BJ, Koh S, Cho HJ, Jin X, Kim BG, Choi JY. High mobility group box 1 in the central nervous system: regeneration hidden beneath inflammation. Neural Regen Res 2025; 20:107-115. [PMID: 38767480 PMCID: PMC11246138 DOI: 10.4103/nrr.nrr-d-23-01964] [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: 11/29/2023] [Revised: 01/31/2024] [Accepted: 03/04/2024] [Indexed: 05/22/2024] Open
Abstract
High-mobility group box 1 was first discovered in the calf thymus as a DNA-binding nuclear protein and has been widely studied in diverse fields, including neurology and neuroscience. High-mobility group box 1 in the extracellular space functions as a pro-inflammatory damage-associated molecular pattern, which has been proven to play an important role in a wide variety of central nervous system disorders such as ischemic stroke, Alzheimer's disease, frontotemporal dementia, Parkinson's disease, multiple sclerosis, epilepsy, and traumatic brain injury. Several drugs that inhibit high-mobility group box 1 as a damage-associated molecular pattern, such as glycyrrhizin, ethyl pyruvate, and neutralizing anti-high-mobility group box 1 antibodies, are commonly used to target high-mobility group box 1 activity in central nervous system disorders. Although it is commonly known for its detrimental inflammatory effect, high-mobility group box 1 has also been shown to have beneficial pro-regenerative roles in central nervous system disorders. In this narrative review, we provide a brief summary of the history of high-mobility group box 1 research and its characterization as a damage-associated molecular pattern, its downstream receptors, and intracellular signaling pathways, how high-mobility group box 1 exerts the repair-favoring roles in general and in the central nervous system, and clues on how to differentiate the pro-regenerative from the pro-inflammatory role. Research targeting high-mobility group box 1 in the central nervous system may benefit from differentiating between the two functions rather than overall suppression of high-mobility group box 1.
Collapse
Affiliation(s)
- Hanki Kim
- Department of Brain Science, Ajou University School of Medicine, Suwon, South Korea
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, South Korea
| | - Bum Jun Kim
- Department of Brain Science, Ajou University School of Medicine, Suwon, South Korea
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, South Korea
| | - Seungyon Koh
- Department of Brain Science, Ajou University School of Medicine, Suwon, South Korea
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, South Korea
- Department of Neurology, Ajou University School of Medicine, Suwon, South Korea
| | - Hyo Jin Cho
- Department of Brain Science, Ajou University School of Medicine, Suwon, South Korea
| | - Xuelian Jin
- Department of Brain Science, Ajou University School of Medicine, Suwon, South Korea
- Department of Geriatrics, The Affiliated Suqian First People’s Hospital of Nanjing Medical University, Suqian, Jiangsu Province, China
| | - Byung Gon Kim
- Department of Brain Science, Ajou University School of Medicine, Suwon, South Korea
- Department of Neurology, Ajou University School of Medicine, Suwon, South Korea
| | - Jun Young Choi
- Department of Brain Science, Ajou University School of Medicine, Suwon, South Korea
- Department of Neurology, Ajou University School of Medicine, Suwon, South Korea
| |
Collapse
|
46
|
Chen X, Lin X. Sodium selenite antagonizes trimethyl tin-induced chicken hepatotoxic hepatitis through the RNS/NF-κB/NLRP3 pathway. Poult Sci 2025; 104:104449. [PMID: 39577171 PMCID: PMC11617673 DOI: 10.1016/j.psj.2024.104449] [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: 08/22/2024] [Revised: 10/10/2024] [Accepted: 10/27/2024] [Indexed: 11/24/2024] Open
Abstract
Trimethyl tin (TMT) is a good stabilizer for plastic products but is also a toxic environmental pollutant. Selenium has good antioxidant and anti-inflammatory properties and has been widely used in the poultry industry. However, it has not been reported whether selenium enrichment can antagonize TMT-induced viral hepatitis in poultry. To fill this gap, AA broiler models exposed to TMT for 42 d were established and fed a Se-enriched diet at the same time. H&E staining showed that selenium could significantly alleviate TMT-induced liver inflammation. Further analysis of the underlying mechanism revealed that TMT induced nitrosation stress (RNS), increased NO content and iNOS expression, which in turn activated the NF-κB/NLRP3 pathway and induced pyroptosis. However, selenium enrichment can reverse this situation, that is, reduce the occurrence of RNS, reduce the degree of pyroptosis, and thus alleviate the occurrence of inflammation. In our study, we demonstrated for the first time that TMT could induce hepatotoxicity other than neurotoxicity in poultry and that selenium could antagonize TMT-induced hepatotoxic hepatitis in chickens through the RNS/NF-κB/NLRP3 pathway.
Collapse
Affiliation(s)
- Xinzhang Chen
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China; Animal Science Faculty of Technology, Northeast Agricultural University, Harbin 150030, PR China
| | - Xu Lin
- Animal Science Faculty of Technology, Northeast Agricultural University, Harbin 150030, PR China.
| |
Collapse
|
47
|
Schiavinato M, Ronanki S, Estruch IM, van den Brink N. Immune response accelerated telomere shortening during early life stage of a passerine bird, the blue tit ( Cyanistes caeruleus). Biol Lett 2025; 21:20240618. [PMID: 39837488 PMCID: PMC11750392 DOI: 10.1098/rsbl.2024.0618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 12/03/2024] [Accepted: 12/03/2024] [Indexed: 01/23/2025] Open
Abstract
Dealing with infections is a daily challenge for wild animals. Empirical data show an increase in reactive oxygen species (ROS) production during immune response. This could have consequences on telomere length, the end parts of linear chromosomes, commonly used as proxy for good health and ageing. Telomere length dynamics may reflect the costs associated with physiological responses. In this study, immune system of blue tit (Cyanistes caeruleus) nestlings was experimentally challenged through a polyinosinic:polycytidylic acid (poly I:C) injection, a synthetic double-stranded RNA that mimics a virus, activating the pathway of immune response triggered via the toll-like receptors 3. This path is known to form ROS downstream. Immune response was quantified by white cell counts in blood, while brain lipoperoxidation has been evaluated as an indicator of oxidative damage. Finally, individuals' telomere length shortening between days 8 and 15 after hatching was measured in erythrocytes. Challenged nestlings showed increased leukocyte number when compared with control (treated with a saline solution), lower brain lipid peroxidation (likely as a result of a compensatory mechanism after oxidative stress burst) and accelerated telomere shortening. These findings support the 'ageing cost of infections pathway' hypothesis, which supposes a role for infections in quick biological ageing.
Collapse
Affiliation(s)
- Matteo Schiavinato
- Cluster of Biomolecular Science, Division of Toxicology, Wageningen University and Research, 6708 WE Wageningen, The Netherlands
| | - Shivani Ronanki
- Cluster of Biomolecular Science, Division of Toxicology, Wageningen University and Research, 6708 WE Wageningen, The Netherlands
| | - Ignacio Miro Estruch
- Cluster of Biomolecular Science, Division of Toxicology, Wageningen University and Research, 6708 WE Wageningen, The Netherlands
| | - Nico van den Brink
- Cluster of Biomolecular Science, Division of Toxicology, Wageningen University and Research, 6708 WE Wageningen, The Netherlands
| |
Collapse
|
48
|
El Gazzar WB, Farag AA, Samir M, Bayoumi H, Youssef HS, Marei YM, Mohamed SK, Marei AM, Abdelfatah RM, Mahmoud MM, Aboelkomsan EAF, Khalfallah EKM, Anwer HM. Berberine chloride loaded nano-PEGylated liposomes attenuates imidacloprid-induced neurotoxicity by inhibiting NLRP3/Caspase-1/GSDMD-mediated pyroptosis. Biofactors 2025; 51:e2107. [PMID: 39074847 DOI: 10.1002/biof.2107] [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: 04/26/2024] [Accepted: 06/25/2024] [Indexed: 07/31/2024]
Abstract
Concerns have been expressed about imidacloprid (IMI), one of the most often used pesticides, and its potential neurotoxicity to non-target organisms. Chronic neuroinflammation is central to the pathology of several neurodegenerative disorders. Hence, exploring the molecular mechanism by which IMI would trigger neuroinflammation is particularly important. This study examined the neurotoxic effects of oral administration of IMI (45 mg/kg/day for 30 days) and the potential neuroprotective effect of berberine (Ber) chloride loaded nano-PEGylated liposomes (Ber-Lip) (10 mg/kg, intravenously every other day for 30 days) using laboratory rat. The histopathological changes, anti-oxidant and oxidative stress markers (GSH, SOD, and MDA), proinflammatory cytokines (IL1β and TNF-α), microglia phenotype markers (CD86 and iNOS for M1; CD163 for M2), the canonical pyroptotic pathway markers (NLRP3, caspase-1, GSDMD, and IL-18) and Alzheimer's disease markers (Neprilysin and beta amyloid [Aβ] deposits) were assessed. Oral administration of IMI resulted in apparent cerebellar histopathological alterations, oxidative stress, predominance of M1 microglia phenotype, significantly upregulated NLRP3, caspase-1, GSDMD, IL-18 and Aβ deposits and significantly decreased Neprilysin expression. Berberine reduced the IMI-induced aberrations in the measured parameters and improved the IMI-induced histopathological and ultrastructure alterations brought on by IMI. This study highlights the IMI neurotoxic effect and its potential contribution to the development of Alzheimer's disease and displayed the neuroprotective effect of Ber-Lip.
Collapse
Affiliation(s)
- Walaa Bayoumie El Gazzar
- Department of Anatomy, Physiology and Biochemistry, Faculty of Medicine, The Hashemite University, Zarqa, Jordan
- Department of Medical Biochemistry and Molecular biology, Faculty of Medicine, Benha University, Benha City, Qalyubia, Egypt
| | - Amina A Farag
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Benha University, Benha City, Qalyubia, Egypt
| | - Mohamed Samir
- Department of Zoonoses, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Sharqia, Egypt
- School of Science, Faculty of Engineering and Science, University of Greenwich, Kent, UK
| | - Heba Bayoumi
- Department of Histology and Cell Biology, Faculty of Medicine, Benha University, Benha City, Egypt
| | - Heba S Youssef
- Department of Physiology, Faculty of Medicine, Benha University, Benha City, Qalyubia, Egypt
| | - Yasmin Mohammed Marei
- Department of Medical Biochemistry and Molecular biology, Faculty of Medicine, Benha University, Benha City, Qalyubia, Egypt
| | - Shimaa K Mohamed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Helwan University, Cairo, Egypt
| | - Azza M Marei
- Department of Zoology, Faculty of Science, Benha University, Benha City, Qalyubia, Egypt
| | - Reham M Abdelfatah
- Department of Pesticides, Faculty of Agriculture, Mansoura University, Mansoura, Egypt
| | | | | | - Eman Kamel M Khalfallah
- Department of Biochemistry, Toxicology and Feed Deficiency, Animal Health Research Institute (AHRI), Agricultural Research Center (ARC), Dokki, Giza, Egypt
| | - Hala Magdy Anwer
- Department of Physiology, Faculty of Medicine, Benha University, Benha City, Qalyubia, Egypt
| |
Collapse
|
49
|
Kulow VA, Roegner K, Labes R, Kasim M, Mathia S, Czopek CS, Berndt N, Becker PN, Ter-Avetisyan G, Luft FC, Enghard P, Hinze C, Klocke J, Eckardt KU, Schmidt-Ott KM, Persson PB, Rosenberger C, Fähling M. Beyond hemoglobin: Critical role of 2,3-bisphosphoglycerate mutase in kidney function and injury. Acta Physiol (Oxf) 2025; 241:e14242. [PMID: 39422260 DOI: 10.1111/apha.14242] [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: 08/14/2024] [Revised: 10/02/2024] [Accepted: 10/03/2024] [Indexed: 10/19/2024]
Abstract
AIM 2,3-bisphosphoglycerate mutase (BPGM) is traditionally recognized for its role in modulating oxygen affinity to hemoglobin in erythrocytes. Recent transcriptomic analyses, however, have indicated a significant upregulation of BPGM in acutely injured murine and human kidneys, suggesting a potential renal function for this enzyme. Here we aim to explore the physiological role of BPGM in the kidney. METHODS A tubular-specific, doxycycline-inducible Bpgm-knockout mouse model was generated. Histological, immunofluorescence, and proteomic analyses were conducted to examine the localization of BPGM expression and the impact of its knockout on kidney structure and function. In vitro studies were performed to investigate the metabolic consequences of Bpgm knockdown under osmotic stress. RESULTS BPGM expression was localized to the distal nephron and was absent in proximal tubules. Inducible knockout of Bpgm resulted in rapid kidney injury within 4 days, characterized by proximal tubular damage and tubulointerstitial fibrosis. Proteomic analyses revealed involvement of BPGM in key metabolic pathways, including glycolysis, oxidative stress response, and inflammation. In vitro, Bpgm knockdown led to enhanced glycolysis, decreased reactive oxygen species elimination capacity under osmotic stress, and increased apoptosis. Furthermore, interactions between nephron segments and immune cells in the kidney suggested a mechanism for propagating stress signals from distal to proximal tubules. CONCLUSION BPGM fulfills critical functions beyond the erythrocyte in maintaining glucose metabolism in the distal nephron. Its absence leads to metabolic imbalances, increased oxidative stress, inflammation, and ultimately kidney injury.
Collapse
Affiliation(s)
- Vera A Kulow
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Medizinische Klinik m.S. Nephrologie und Internistische Intensivmedizin (CCM), Berlin, Germany
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institut für Translationale Physiologie (CCM), Berlin, Germany
| | - Kameliya Roegner
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institut für Translationale Physiologie (CCM), Berlin, Germany
| | - Robert Labes
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institut für Translationale Physiologie (CCM), Berlin, Germany
| | - Mumtaz Kasim
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institut für Translationale Physiologie (CCM), Berlin, Germany
| | - Susanne Mathia
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Medizinische Klinik m.S. Nephrologie und Internistische Intensivmedizin (CCM), Berlin, Germany
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institut für Translationale Physiologie (CCM), Berlin, Germany
| | - Claudia S Czopek
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institut für Translationale Physiologie (CCM), Berlin, Germany
| | - Nikolaus Berndt
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), Nuthetal, Germany
- Deutsches Herzzentrum der Charité (DHZC), Institute of Computer-assisted Cardiovascular Medicine, Berlin, Germany
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Philipp N Becker
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institut für Translationale Physiologie (CCM), Berlin, Germany
| | - Gohar Ter-Avetisyan
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institut für Translationale Physiologie (CCM), Berlin, Germany
| | - Friedrich C Luft
- Experimental and Clinical Research Center (ECRC), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Philipp Enghard
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Medizinische Klinik m.S. Nephrologie und Internistische Intensivmedizin (CCM), Berlin, Germany
| | - Christian Hinze
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Medizinische Klinik m.S. Nephrologie und Internistische Intensivmedizin (CCM), Berlin, Germany
- Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | - Jan Klocke
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Medizinische Klinik m.S. Nephrologie und Internistische Intensivmedizin (CCM), Berlin, Germany
| | - Kai-Uwe Eckardt
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Medizinische Klinik m.S. Nephrologie und Internistische Intensivmedizin (CCM), Berlin, Germany
| | - Kai M Schmidt-Ott
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Medizinische Klinik m.S. Nephrologie und Internistische Intensivmedizin (CCM), Berlin, Germany
- Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | - Pontus B Persson
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institut für Translationale Physiologie (CCM), Berlin, Germany
| | - Christian Rosenberger
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Medizinische Klinik m.S. Nephrologie und Internistische Intensivmedizin (CCM), Berlin, Germany
| | - Michael Fähling
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institut für Translationale Physiologie (CCM), Berlin, Germany
| |
Collapse
|
50
|
Vena W, Pigni S, Betella N, Navarra A, Mirani M, Mazziotti G, Lania AG, Bossi AC. COVID-19 vaccines and blood glucose control: Friend or foe? Hum Vaccin Immunother 2024; 20:2363068. [PMID: 38860457 PMCID: PMC11178329 DOI: 10.1080/21645515.2024.2363068] [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: 02/20/2024] [Accepted: 05/30/2024] [Indexed: 06/12/2024] Open
Abstract
PURPOSE To overview the recent literature regarding the relationship between COVID-19 vaccines and glycemic control. METHODS Data were extracted from text and tables of all available articles published up to September 2023 in PubMed Database describing glucose homeostasis data in subjects exposed to COVID-19 vaccines, focusing on patients with diabetes mellitus (DM). RESULTS It is debated if the immune system impairment observed in diabetic patients makes them susceptible to lower efficacy of vaccines, but evidence suggests a possible improvement in immune response in those with good glycemic control. Despite their proven protective role lowering infection rates and disease severity, COVID-19 vaccines can result in diabetic ketoacidosis, new-onset diabetes, or episodes of hyper- or hypoglycemia. CONCLUSIONS Evidence with COVID-19 vaccines highlights the strong relationship existing between DM and immune system function. Clinicians should strive to achieve optimal glucose control before vaccination and promptly manage possible glucose homeostasis derangement following vaccine exposure.
Collapse
Affiliation(s)
- Walter Vena
- Department of Biomedical Sciences, Humanitas University, Milan, Pieve Emanuele, Italy
- Diabetes Center, Humanitas Gavazzeni Institute, Bergamo, Italy
| | - Stella Pigni
- Department of Biomedical Sciences, Humanitas University, Milan, Pieve Emanuele, Italy
- Endocrinology, Diabetology and Medical Andrology Unit, IRCCS Humanitas Research Hospital, Milan, Rozzano, Italy
| | | | | | - Marco Mirani
- Endocrinology, Diabetology and Medical Andrology Unit, IRCCS Humanitas Research Hospital, Milan, Rozzano, Italy
| | - Gherardo Mazziotti
- Department of Biomedical Sciences, Humanitas University, Milan, Pieve Emanuele, Italy
- Endocrinology, Diabetology and Medical Andrology Unit, IRCCS Humanitas Research Hospital, Milan, Rozzano, Italy
| | - Andrea G. Lania
- Department of Biomedical Sciences, Humanitas University, Milan, Pieve Emanuele, Italy
- Endocrinology, Diabetology and Medical Andrology Unit, IRCCS Humanitas Research Hospital, Milan, Rozzano, Italy
| | | |
Collapse
|