1
|
Cheng F, Li Y, Deng K, Zhang X, Sun W, Yang X, Zhang X, Wang C. Associations between phthalate metabolites and two novel systemic inflammatory indexes: a cross-sectional analysis of NHANES data. Ann Med 2025; 57:2496411. [PMID: 40272105 PMCID: PMC12024508 DOI: 10.1080/07853890.2025.2496411] [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: 12/20/2024] [Revised: 03/17/2025] [Accepted: 04/14/2025] [Indexed: 04/25/2025] Open
Abstract
BACKGROUND The potentially risky effects of metabolites of phthalates (mPAEs) on inflammation and immune function have attracted much attention in recent years. However, direct studies on the relationship between these metabolites and the systemic immune inflammatory index (SII) and systemic inflammatory response index (SIRI) are limited. METHODS This cross-sectional study used generalized linear regression models (GLM), restricted cubic splines (RCS), weighted quantile sum (WQS), and Bayesian kernel-machine regression (BKMR) to analyze data from 2,763 U.S. adults aged between 20 and 80 years, obtained from the U.S. National Health and Nutrition Examination Survey (NHANES) conducted between 2013 and 2018. The study aimed to investigate the relationship between urine samples of nine mPAEs and levels of SII/SIRI in a single, nonlinear, and mixed relationship and explored the robustness of the findings under single and mixed effects using two sensitivity analyses for completeness. In addition, the effects of six variables (age, sex, BMI, the percentage of total daily energy intake from ultra-processed foods (UPFs), total vegetable intake, and dietary supplements) on the association results were explored through subgroup analyses to identify potentially important confounders. RESULTS In single exposure analyses, mono-n-butyl phthalate (MnBP), mono-ethyl phthalate (MEP), and monobenzyl phthalate (MBzP) were positively associated with SII/SIRI. The findings from the two mixed exposure models demonstrated a positive association between the collective concentrations of mPAEs and levels of SII/SIRI, with MBzP being identified as a significant contributor to the urinary levels of mPAEs. The subgroup analysis results of the effects of single and mixed exposures show that the association between mPAEs and SII/SIRI is more significant in females, overweight/obese populations, young/middle-aged populations, and populations with high levels of intake of UPFs. CONCLUSION Positive associations were identified between mPAEs and SII/SIRI. MBzP was determined to have the most significant impact. The association between mPAEs and SII/SIRI is significantly influenced by female groups, young and middle-aged populations, overweight and obese individuals, as well as those with a higher intake of UPFs.
Collapse
Affiliation(s)
- Fangyu Cheng
- School of Public Health, Shandong Second Medical University, Weifang, Shandong, China
| | - Yueyuan Li
- School of Public Health, Shandong Second Medical University, Weifang, Shandong, China
| | - Kai Deng
- School of Public Health, Shandong Second Medical University, Weifang, Shandong, China
| | - Xinyu Zhang
- School of Public Health, Shandong Second Medical University, Weifang, Shandong, China
| | - Wenxue Sun
- School of Public Health, Shandong Second Medical University, Weifang, Shandong, China
| | - Xin Yang
- School of Public Health, Shandong Second Medical University, Weifang, Shandong, China
| | - Xiaofang Zhang
- School of Public Health, Shandong Second Medical University, Weifang, Shandong, China
| | - Chunping Wang
- School of Public Health, Shandong Second Medical University, Weifang, Shandong, China
| |
Collapse
|
2
|
Tian W, Ju J, Guan B, Wang T, Zhang J, Song L, Xu H. Role of hyperhomocysteinemia in atherosclerosis: from bench to bedside. Ann Med 2025; 57:2457527. [PMID: 39898976 PMCID: PMC11792134 DOI: 10.1080/07853890.2025.2457527] [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: 07/20/2024] [Revised: 01/07/2025] [Accepted: 01/08/2025] [Indexed: 02/04/2025] Open
Abstract
BACKGROUND Atherosclerosis is a leading cause of global mortality, driven by complex interactions between genetic, metabolic, and environmental factors. Among these, hyperhomocysteinemia (HHcy) has emerged as a significant and modifiable risk factor, contributing to endothelial dysfunction, oxidative stress, and vascular inflammation. Despite increasing recognition of its role in atherogenesis, the precise mechanisms and clinical implications of HHcy remain incompletely understood, necessitating a comprehensive review to connect recent mechanistic insights with practical applications. METHODS We analyzed the various mechanisms whereby HHcy accelerates the progression of atherosclerosis, and conducted a comprehensive review of publications in the fields of HHcy and atherosclerosis. RESULTS HHcy promotes atherosclerosis through several mechanisms, including inflammation, oxidative stress, epigenetic modification, and lipoprotein metabolism alteration. Moreover, this discussion extends to current strategies for the prevention and clinical management of HHcy-induced atherosclerosis. CONCLUSION This review consolidates and elucidates the latest advancements and insights into the role of HHcy in atherosclerosis. The comprehensive narrative connects fundamental research with clinical applications. Contemporary studies highlight the complex interplay between HHcy and atherosclerosis, establishing HHcy as not only a contributing risk factor but also an accelerator of various atherogenic processes.
Collapse
Affiliation(s)
- Wende Tian
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing China
- Graduate School, China Academy of Chinese Medical Sciences, Beijing China
| | - Jianqing Ju
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing China
| | - Baoyi Guan
- Department of Internal Medicine-Cardiovascular, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Tongxin Wang
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing China
- Graduate School, China Academy of Chinese Medical Sciences, Beijing China
| | - Jiqian Zhang
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Luxia Song
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing China
| | - Hao Xu
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing China
| |
Collapse
|
3
|
Ban G, Chen Y, Liang Y, Wang X, Ding D, Liu R, Jia J, Zhao R, Wang C, Li N. Exploring the efficacy and constraints of platinum nanoparticles as adjuvant therapy in silicosis management. Drug Deliv 2025; 32:2445257. [PMID: 39803920 PMCID: PMC11730774 DOI: 10.1080/10717544.2024.2445257] [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: 09/02/2024] [Revised: 11/19/2024] [Accepted: 12/10/2024] [Indexed: 01/16/2025] Open
Abstract
Silicosis represents a formidable occupational lung pathology precipitated by the pulmonary assimilation of respirable crystalline silica particulates. This condition engenders a cascade of cellular oxidative stress via the activation of bioavailable silica, culminating in the generation of reactive oxygen species (ROS). Such oxidative mechanisms lead to irrevocable pulmonary impairment. Contemporary scholarly examinations have underscored the substantial antioxidative efficacy of platinum nanoparticles (PtNPs), postulating their utility as an adjunct therapeutic modality in silicosis management. The physicochemical interaction between PtNPs and silica demonstrates a propensity for adsorption, thereby facilitating the amelioration and subsequent pulmonary clearance of silica aggregates. In addition to their detoxifying attributes, PtNPs exhibit pronounced anti-inflammatory and antioxidative activities, which can neutralize ROS and inhibit macrophage-mediated inflammatory processes. Such attributes are instrumental in attenuating inflammatory responses and forestalling subsequent lung tissue damage. This discourse delineates the interplay between ROS and PtNPs, the pathogenesis of silicosis and its progression to pulmonary fibrosis, and critically evaluates the potential adjunct role of PtNPs in the therapeutic landscape of silicosis, alongside a contemplation of the inherent limitations associated with PtNPs application in this context.
Collapse
Affiliation(s)
- Ge Ban
- School of Intelligent Medical Engineering, Sanquan College of Xinxiang Medical University, Xinxiang, China
| | - Yuanjie Chen
- School of Intelligent Medical Engineering, Sanquan College of Xinxiang Medical University, Xinxiang, China
- Clinical School, Sanquan College of Xinxiang Medical University, Xinxiang, China
| | - Yingbing Liang
- Department of Chemistry and Biotechnology, Graduate School of Engineering Tottori University, Tottori, Japan
| | - Xiaona Wang
- School of Intelligent Medical Engineering, Sanquan College of Xinxiang Medical University, Xinxiang, China
| | - Dan Ding
- School of Intelligent Medical Engineering, Sanquan College of Xinxiang Medical University, Xinxiang, China
| | - Rui Liu
- School of Life Sciences and Biotechnology, Sanquan College of Xinxiang Medical University, China
| | - Jingjing Jia
- School of Intelligent Medical Engineering, Sanquan College of Xinxiang Medical University, Xinxiang, China
| | - Ran Zhao
- School of Intelligent Medical Engineering, Sanquan College of Xinxiang Medical University, Xinxiang, China
| | - Chenxia Wang
- Department of Respiratory Medicine, People’s Hospital of Huojia County, Xinxiang, China
| | - Na Li
- College of Pharmacy, Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
4
|
Ahmed AKM, Beshr EAM, Salem IM, Ahmed OAA, Ibrahim TS, Elsayed Abouzed DE, Mahmoud AM, Mohamed MFA. Novel chalcone candidates as potential in vitro and in vivo anti-inflammatory agents: Synthesis, in silico docking, multitarget bioevaluation and molecular dynamic simulation. Bioorg Chem 2025; 161:108540. [PMID: 40306188 DOI: 10.1016/j.bioorg.2025.108540] [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: 02/16/2025] [Revised: 04/24/2025] [Accepted: 04/25/2025] [Indexed: 05/02/2025]
Abstract
Managing inflammation with the commonly used nonsteroidal anti-inflammatory drugs (NSAIDs) represents a critical challenge in modern medicine because of its strong influence on the cyclooxygenase-1 (COX-1) enzyme might induce substantial adverse effects. Therefore, there is urgent necessity for the exploration of safer alternatives, particularly cyclooxygenase-2 (COX-2) inhibitors. This study aimed to address this issue through the synthesis and evaluation of 19 new chalcone derivatives (2a-m and 4a-f) for their in vitro anti-inflammatory activity against various biotargets including iNOS, COX-2, 5-LOX, PGE2, and TNFα. Moreover, these compounds showed moderate to strong anti-inflammatory activity in the carrageenan rat paw edema test. Compounds 2a, 2f, 2 h, 2 m, and 4b are promising candidates for the treatment of inflammatory diseases. In particular, compound 4b was demonstrated to be the most effective derivative as a nitric oxide release inhibitor, exhibiting a 61.7 % inhibition rate. It exhibited substantial selectivity for COX-2 (IC50 = 1.933 μM) compared to COX-1 (IC50 = 5.526 μM). Compound 4b exhibited notable inhibitory activity against 5-LOX (IC50 = 2.112 μM) and demonstrated considerable inhibitory activity against iNOS, PGE2, and TNF-α biotargets in LPS-stimulated RAW cells, with IC50 values of 114.18, 37.13, and 58.15 nM, respectively. The in vivo anti-inflammatory effects demonstrated the significant efficacy of compound 4b, as evidenced by a notable edema inhibition rate of 37.05 %, along with minimal ulcerogenic activity observed in the histopathological findings. In silico experiments demonstrated that the intermolecular contacts of the most active chemical 4b with the biotargets COX-2, 5-LOX, and iNOS were analyzed by docking, revealing significant binding interactions. The stability of the interactions between compound 4b and the targets COX-2, 5-LOX, and iNOS was assessed using a standard 100 ns atomistic dynamic simulation method. Various parameters derived from MD simulation trajectories were adjusted and validated to confirm the stability of the generated complexes under dynamic settings. Ultimately, compound 4b exhibited favorable physicochemical properties and satisfactory drug-likeness, indicating its potential as an oral anti-inflammatory agent, warranting additional structure-activity relationship investigation and optimization.
Collapse
Affiliation(s)
- Alshimaa Kh M Ahmed
- Department of Medicinal Chemistry, Faculty of Pharmacy, Minia University, Minia 61519, Egypt
| | - Eman A M Beshr
- Department of Medicinal Chemistry, Faculty of Pharmacy, Minia University, Minia 61519, Egypt; Medicinal Chemistry Department, Faculty of Pharmacy, Minia National University, New Minia, Egypt.
| | - Ibrahim M Salem
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Sphinx University, New Assiut City, Assiut 71515, Egypt
| | - Osama A A Ahmed
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, 21589 Jeddah, Saudi Arabia; Mohamed Saeed Tamer Chair for Pharmaceutical Industries, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Tarek S Ibrahim
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Deiaa E Elsayed Abouzed
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Sohag University, Sohag 82524, Egypt
| | - Ahmed Mostafa Mahmoud
- Department of Physiology, Faculty of Medicine, Sohag University, Sohag, Egypt; Department of Basic Medical Sciences, Aqaba Medical Sciences University, Aqaba, Jordan
| | - Mamdouh F A Mohamed
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Sohag University, 82524 Sohag, Egypt; Department of Pharmaceutical Chemistry, Faculty of Pharmacy, New Valley University, New Valley, 72511, Egypt.
| |
Collapse
|
5
|
Lin C, Li ZS, Dong ZW, Wu XY, Ye M, Li K, Jin Z, Wang W, Tang YZ. Discovery and optimization of ibuprofen derivatives as the NF-κB/iNOS pathway inhibitors for the treatment of ulcerative colitis. Bioorg Chem 2025; 161:108506. [PMID: 40311243 DOI: 10.1016/j.bioorg.2025.108506] [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: 02/10/2025] [Revised: 04/01/2025] [Accepted: 04/21/2025] [Indexed: 05/03/2025]
Abstract
In this study, a series of novel ibuprofen (Ibu) hybrid molecules with aminothiazole heterocycles were designed, synthesized and evaluated for their anti-inflammatory potency in vitro and in vivo. Among all these derivatives, compounds 6 and 8 effectively inhibited the production of NO (with 87 %, 79 % NO-inhibitory rates, respectively) with minimal cytotoxic effect in RAW 264.7 macrophages. Anti-inflammatory mechanism studies revealed that representative compound 6 dose-dependently inhibited pro-inflammatory cytokines by blocking the activation of NF-κB signaling pathway in LPS stimulated RAW 264.7 macrophages. In vivo experiments showed that 10 mg/kg compound 6 had a good improvement effect in DSS-induced mouse acute colitis compared to Ibu. Our findings will provide new insights into the development of new drugs with anti-inflammatory functions.
Collapse
Affiliation(s)
- Chao Lin
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Zhong-Sheng Li
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Zhan-Wei Dong
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Xiao-Yi Wu
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Min Ye
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Ke Li
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Zhen Jin
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Wei Wang
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China.
| | - You-Zhi Tang
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China.
| |
Collapse
|
6
|
Liu H, Xue H, Guo Q, Xue X, Yang L, Zhao K, Liu Y. Ferroptosis meets inflammation: A new frontier in cancer therapy. Cancer Lett 2025; 620:217696. [PMID: 40189012 DOI: 10.1016/j.canlet.2025.217696] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Revised: 03/26/2025] [Accepted: 04/03/2025] [Indexed: 04/10/2025]
Abstract
Ferroptosis, an iron-dependent form of regulated cell death driven by lipid peroxidation, has emerged as a critical player in cancer pathogenesis. Concurrently, inflammation, a key biological response to tissue injury or infection, significantly influences cancer development and progression. The interplay between ferroptosis and inflammation represents a promising yet underexplored area of research. This review synthesizes recent advances in understanding the molecular mechanisms governing their interaction, emphasizing how ferroptosis triggers inflammatory responses and how inflammatory mediators, such as TNF-α, regulate ferroptosis through iron metabolism and lipid peroxidation pathways. Key molecular targets within the ferroptosis-inflammation axis, including GPX4, ACSL4, and the NF-κB signaling pathway, offer therapeutic potential for cancer treatment. By modulating these targets, it may be possible to enhance ferroptosis and fine-tune inflammatory responses, thereby improving therapeutic outcomes. Additionally, this review explores the broader implications of targeting the ferroptosis-inflammation interplay in disease treatment, highlighting opportunities for developing innovative strategies to combat cancer. By bridging the gap in current knowledge, this review provides a comprehensive resource for researchers and clinicians, offering insights into the therapeutic potential of this intricate biological relationship.
Collapse
Affiliation(s)
- Hu Liu
- Department of Oncology Surgery, Shanghai Mengchao Hospital, Shanghai University, Shanghai, 202800, China
| | - Hui Xue
- Department of Oncology Surgery, Shanghai Mengchao Hospital, Shanghai University, Shanghai, 202800, China
| | - Qian Guo
- Department of Rhinology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Xutong Xue
- Boston Children's Hospital, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Lixue Yang
- Department of Oncology Surgery, Shanghai Mengchao Hospital, Shanghai University, Shanghai, 202800, China.
| | - Kaijun Zhao
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China.
| | - Yu'e Liu
- Boston Children's Hospital, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, 02115, USA; Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China.
| |
Collapse
|
7
|
Vivesh V, Kumari P, Singh P. A rationale approach in developing proline based small peptide as U-shaped analogues of arachidonic acid: Dual inhibitors of cyclooxygenase-2 and 5-lipoxygenase for developing anti-inflammatory agents. Bioorg Chem 2025; 160:108452. [PMID: 40253762 DOI: 10.1016/j.bioorg.2025.108452] [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: 01/28/2025] [Revised: 03/29/2025] [Accepted: 04/07/2025] [Indexed: 04/22/2025]
Abstract
Guided by the molecular modelling studies, here, we report the development of small molecules containing Pro-Pro, Gly-Pro-Pro ester motifs as dual inhibitors of COX-2 and 5-LOX enzymes. The synthesized compounds exhibited potent inhibitory activities against COX-2 and 5-LOX, with IC50 values in nanomolar range. Lineweaver-Burk plot analysis revealed that these molecules act as competitive inhibitors of COX-2 and 5-LOX. In vivo studies using the HET-CAM assay indicated that the compounds exhibit moderate to good anti-inflammatory effects. Hence, taking into account the physicochemical properties, including aqueous solubility, binding affinity to HSA and stability in blood plasma and liver microsomes, anti-inflammatory agents targeting dual pathways of arachidonic acid metabolism are identified.
Collapse
Affiliation(s)
- Vivesh Vivesh
- Department of Chemistry, Guru Nanak Dev University Amritsar, 143005, India
| | - Priya Kumari
- Department of Chemistry, Guru Nanak Dev University Amritsar, 143005, India
| | - Palwinder Singh
- Department of Chemistry, Guru Nanak Dev University Amritsar, 143005, India.
| |
Collapse
|
8
|
Sui Q, Zhu C, Shi S, Xu J, Zhang J, Wang A, Chen P, Liang G, Zhang Y. Ganoderic acid A: an in-depth review of pharmacological effects and molecular docking analysis. JOURNAL OF ETHNOPHARMACOLOGY 2025; 349:119868. [PMID: 40316150 DOI: 10.1016/j.jep.2025.119868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 03/24/2025] [Accepted: 04/22/2025] [Indexed: 05/04/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Ganoderic acid A (GAA, C30H44O7) is one of the most abundant and active components of Ganoderic acids (GAs). GAs are highly oxidized tetracyclic triterpenoid compounds mainly derived from Ganoderma lucidum (Curtis) P. Karst (Chinese: ). GAA is primarily isolated from the fruiting body of Ganoderma lucidum. Modern pharmacological investigations have established the broad pharmacological effects of GAA, highlighting its notable influence on managing various conditions, including inflammatory diseases, neurodegenerative diseases, and cancer. This review provides a comprehensive summary of GAA's pharmacological activities. MATERIAL AND METHODS The literature in this review were searched in PubMed and China National Knowledge Infrastructure (CNKI) using the keywords "Ganoderic acid A″, "Pharmacology" and "Pharmacokinetics". The literature cited in this review dates from 2000 to 2024. RESULTS According to the data, GAA exerts anti-inflammatory, antioxidant, antitumor, neuropsychopharmacological, hepatoprotective, cardiovascular, renoprotective, and lung protective effects by regulating a variety of signal transduction pathways, such as nuclear factor kappa-B (NF-κB), Janus kinase/signal transducer and activator of transcription (JAK/STAT), Toll-like receptor 4 (TLR4), nuclear factor erythroid 2-related factor-2 (Nrf2), phosphoinositide-3-kinase (PI3K)/AKT, mammalian target of rapamycin (mTOR), mitogen-activated protein kinase (MAPK), and Notch. Given its promising pharmacological activity, GAA holds excellent potential for treating human diseases. The pharmacokinetic properties of GAA have also been reviewed, revealing low bioavailability but high absorption and elimination rates. In addition, network pharmacology and molecular docking analyses verified that GAA plays a role in multiple diseases through MAPK3, tumor necrosis factor (TNF), caspase-3 (CASP3), peroxisome proliferator-activated receptor gamma (PPARG), and β-catenin (CTNNB1) signaling pathways. CONCLUSION GAA plays a pivotal role in various pathological and physiological processes, boasting broad application prospects. Furthermore, the network pharmacological results reveal the mechanisms of GAA in the treatment of multiple diseases. In the future, it is necessary to conduct further experiments to elucidate its specific mechanism of action, thus laying the foundation for the scientific utilization of GAA.
Collapse
Affiliation(s)
- Qi Sui
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, 310012, China; Zhejiang TCM Key Laboratory of Pharmacology and Translational Research of Natural Products, Hangzhou Medical College, Hangzhou, Zhejiang, 310012, China
| | - Chengkai Zhu
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, 310012, China; Zhejiang TCM Key Laboratory of Pharmacology and Translational Research of Natural Products, Hangzhou Medical College, Hangzhou, Zhejiang, 310012, China
| | - Sha Shi
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, 310012, China; Zhejiang TCM Key Laboratory of Pharmacology and Translational Research of Natural Products, Hangzhou Medical College, Hangzhou, Zhejiang, 310012, China
| | - Jiaqi Xu
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, 310012, China; Zhejiang TCM Key Laboratory of Pharmacology and Translational Research of Natural Products, Hangzhou Medical College, Hangzhou, Zhejiang, 310012, China
| | - Jingnan Zhang
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, 310012, China; Zhejiang TCM Key Laboratory of Pharmacology and Translational Research of Natural Products, Hangzhou Medical College, Hangzhou, Zhejiang, 310012, China
| | - Ao Wang
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, 310012, China; Department of Pharmacy, School of Medicine, Hangzhou City University, 50 Huzhou Rd, Hangzhou, Zhejiang, 310015, China; Zhejiang TCM Key Laboratory of Pharmacology and Translational Research of Natural Products, Hangzhou Medical College, Hangzhou, Zhejiang, 310012, China; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Peng Chen
- Department of Pharmacy, School of Medicine, Hangzhou City University, 50 Huzhou Rd, Hangzhou, Zhejiang, 310015, China.
| | - Guang Liang
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, 310012, China; Zhejiang TCM Key Laboratory of Pharmacology and Translational Research of Natural Products, Hangzhou Medical College, Hangzhou, Zhejiang, 310012, China; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China.
| | - Yi Zhang
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, 310012, China; Zhejiang TCM Key Laboratory of Pharmacology and Translational Research of Natural Products, Hangzhou Medical College, Hangzhou, Zhejiang, 310012, China.
| |
Collapse
|
9
|
Kumar D, Ahmed M, Andrabi NI, Singh CP, Saroch D, Bharitkar YP, Kour G, Madishetti S, Bhagat A, Shukla SK, Ahmed Z. Anti-inflammatory and anti-oxidant potential of dispiro-indanedione hybrid of parthenin via regulating Nrf2 and NF-κB/MAPK pathways. Eur J Pharmacol 2025; 996:177547. [PMID: 40154568 DOI: 10.1016/j.ejphar.2025.177547] [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: 12/17/2024] [Revised: 02/17/2025] [Accepted: 03/24/2025] [Indexed: 04/01/2025]
Abstract
Steroidal and non-steroidal anti-inflammatory drugs are widely used for treating a spectrum of inflammatory conditions; however, their systemic adverse effects hinder their usage. Therefore, alternate therapeutic strategies are required to treat inflammatory disorders. Parthenin, a lactone derived from Parthenium hysterophorus, has demonstrated anti-inflammatory activity; however, its toxic nature limits its application. We proposed modifications of parthenin to enhance its efficacy while reducing toxicity. In this context, we screened parthenin derivatives for anti-inflammatory efficacy and identified dispiro-indanedione hybrid of parthenin (DIHP) as a potent anti-inflammatory agent. Macrophages were pre-treated with DIHP followed by LPS stimulation to evaluate the in-vitro anti-inflammatory and anti-oxidant activity. We assessed in-vivo anti-inflammatory effect of DIHP in carrageenan-induced paw edema and LPS-induced sepsis model. Our findings showed that DIHP exerts negligible effect on cell viability, effectively attenuates the production of inflammatory markers (NO, TNF-α, IL-6 &IL-1β) and down-regulates NF-κB, MAPK pathways in in-vitro and in-vivo system. Additionally, DIHP inhibited LPS-induced generation of prostaglandin E2, leukotriene B4, ROS and upregulated the expression of superoxide dismutase, catalase, nuclear factor-E2-related factor 2 and peroxisome proliferator-activated receptor gamma. Furthermore, DIHP effectively reduced carrageenan-induced paw edema and curtailed the levels of liver, and kidney damage markers (AST, ALT, CRE, and BUN), protected the lung, liver and kidney against pathological damage and enhanced the survival rate in LPS-challenged mice. DIHP demonstrated comparable efficacy to dexamethasone in reducing inflammatory markers. In conclusion, our study strongly suggests that DIHP curtailed inflammation and oxidative stress by down regulating NF-κB and MAPK pathways and enhanced anti-oxidant response.
Collapse
Affiliation(s)
- Diljeet Kumar
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu, 180001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Manzoor Ahmed
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu, 180001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Nusrit Iqbal Andrabi
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu, 180001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Chetan Paul Singh
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India; Natural Products & Medicinal Chemistry Division, CSIR-Indian Indian Institute of Integrative Medicine, Canal Road, Jammu, 180001, India
| | - Diksha Saroch
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu, 180001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Yogesh P Bharitkar
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India; Natural Products & Medicinal Chemistry Division, CSIR-Indian Indian Institute of Integrative Medicine, Canal Road, Jammu, 180001, India
| | - Gurleen Kour
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu, 180001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Sreedhar Madishetti
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu, 180001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Asha Bhagat
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu, 180001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Sanket K Shukla
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu, 180001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| | - Zabeer Ahmed
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu, 180001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
10
|
Yang H, Liu M, Song S, Xu Q, Lee J, Sun J, Xue S, Sun X, Che C. HIF-1α Promotes Inflammatory Responses in Aspergillus Fumigatus Keratitis by Activating Pyroptosis Through Caspase-8/GSDMD Pathway. Invest Ophthalmol Vis Sci 2025; 66:32. [PMID: 40492985 DOI: 10.1167/iovs.66.6.32] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2025] Open
Abstract
Purpose This research was designed to explore the expression patterns and functional significance of hypoxia-inducible factor-1α (HIF-1α) in the inflammatory response associated with Aspergillus fumigatus (A. fumigatus) keratitis. Methods Mouse models of A. fumigatus keratitis were created by scraping the corneal epithelium and applying A. fumigatus on the corneal surface. In the in vitro experiments, human corneal epithelial cells (HCECs) and THP-1 macrophages stimulated by A. fumigatus were used to investigate the cellular responses. HIF-1α was inhibited using LW6. Western blot, immunofluorescence, and quantitative reverse transcription polymerase chain reaction (qRT-PCR) were performed to assess the expression levels of HIF-1α in A. fumigatus keratitis. The inflammatory response was evaluated using clinical scoring, corneal thickness measurements, hematoxylin and eosin (H&E) staining, corneal fluorescein sodium staining, and a cell scratch test. The polarization of macrophages was determined using flow cytometry. The molecular mechanisms of HIF-1α were assessed by qRT-PCR and Western blot. Results In A. fumigatus keratitis, the expression of HIF-1α was significantly increased at both the mRNA and protein levels. Compared with the controls, HIF-1α inhibitor accelerated corneal epithelial repair, reduced the infiltration of macrophages, induced shift in macrophage polarization, and attenuated the inflammatory response. HIF-1α exerts a pro-inflammatory effect in A. fumigatus keratitis by modulating the expression of inflammatory mediators and engaging in pyroptosis via the caspase-8/GSDMD signaling pathway. Conclusions In conclusion, HIF-1α promotes A. fumigatus keratitis by inhibiting corneal epithelial repair and promoting inflammation, leading to increased severity of the disease.
Collapse
MESH Headings
- Hypoxia-Inducible Factor 1, alpha Subunit/genetics
- Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Animals
- Aspergillus fumigatus
- Mice
- Keratitis/metabolism
- Keratitis/microbiology
- Keratitis/pathology
- Aspergillosis/metabolism
- Aspergillosis/microbiology
- Aspergillosis/pathology
- Eye Infections, Fungal/metabolism
- Eye Infections, Fungal/microbiology
- Eye Infections, Fungal/pathology
- Disease Models, Animal
- Humans
- Pyroptosis/physiology
- Blotting, Western
- Epithelium, Corneal/metabolism
- Epithelium, Corneal/pathology
- Caspase 8/metabolism
- Signal Transduction
- Mice, Inbred C57BL
- Cells, Cultured
- Real-Time Polymerase Chain Reaction
- Flow Cytometry
Collapse
Affiliation(s)
- Hua Yang
- Department of Ophthalmology, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Mengzhu Liu
- Department of Ophthalmology, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Shiqi Song
- Department of Ophthalmology, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Qiang Xu
- Department of Ophthalmology, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jieun Lee
- Department of Ophthalmology, School of Medicine, Pusan National University, Yangsan, Korea
| | - Jintao Sun
- Department of Ophthalmology, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Shasha Xue
- Department of Ophthalmology, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiaoyan Sun
- Department of Ophthalmology, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Chengye Che
- Department of Ophthalmology, the Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
11
|
Askarizadeh F, Butler AE, Kesharwani P, Sahebkar A. Regulatory effect of curcumin on CD40:CD40L interaction and therapeutic implications. Food Chem Toxicol 2025; 200:115369. [PMID: 40043936 DOI: 10.1016/j.fct.2025.115369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 01/24/2025] [Accepted: 03/02/2025] [Indexed: 04/21/2025]
Abstract
Natural compounds have garnered significant attention as potential therapeutic agents due to their inherent properties. Their notable qualities, including safety, efficacy, favorable pharmacokinetic properties, and heightened effectiveness against certain diseases, particularly inflammatory conditions, make them particularly appealing. Among these compounds, curcumin has attracted considerable interest for its unique therapeutic properties and has therefore been extensively studied as a potential therapeutic agent for treating various diseases. Curcumin exhibits diverse anti-inflammatory, antioxidant, and antimicrobial effects. Curcumin's immune system regulatory ability has made it a promising compound for treatment of various inflammatory diseases, such as psoriasis, atherosclerosis, asthma, colitis, IBD, and arthritis. Among the signaling pathways implicated in these conditions, the CD40 receptor together with its ligand, CD40L, are recognized as central players. Studies have demonstrated that the interaction between CD40 and CD40L interaction acts as the primary mediator of the immune response in inflammatory diseases. Numerous studies have explored the impact of curcumin on the CD40:CD40L pathway, highlighting its regulatory effects on this inflammatory pathway and its potential therapeutic use in related inflammatory conditions. In this review, we will consider the evidence concerning curcumin's modulatory effects in inflammatory disease and its potential therapeutic role in regulating the CD40:CD40L pathway.
Collapse
Affiliation(s)
- Fatemeh Askarizadeh
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India.
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Institute of Medical and Technical Sciences, Saveetha Medical College and Hospitals, Saveetha University, Chennai, India; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
12
|
Wang J, Miao Z, Gao Y, Xie Z, Liu M, Zou W. Formyl peptide receptor 2: a potential therapeutic target for inflammation-related diseases. Pharmacol Rep 2025; 77:593-609. [PMID: 40102363 DOI: 10.1007/s43440-025-00704-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 01/23/2025] [Accepted: 01/23/2025] [Indexed: 03/20/2025]
Abstract
Formyl peptide receptor 2 (FPR2) is a G protein-coupled receptor with seven transmembrane domains, widely distributed in human cells. It plays a crucial role in inflammation-related diseases. Known for its "double-edged sword" nature, FPR2 can bind a variety of exogenous and endogenous ligands, mediating both pro-inflammatory and anti-inflammatory responses in tissues such as eyes, liver, joints, lungs, nerves, and blood vessels. FPR2's bioactivities are regulated by a complex network of genes and signaling pathways. However, the precise regulatory mechanisms governing its functions in different inflammatory conditions are still not well understood. This review summarizes the FPR2's activities in various inflammation-related diseases and looks into its potential as a therapeutic target. This review highlights recent advances in developing exogenous agonists for FPR2 and discusses receptor expression across species to support nonclinical research. Overall, this review aims to clarify FPR2's role in inflammation and provide insights for the development of new drugs against inflammatory diseases.
Collapse
Affiliation(s)
- Jiaying Wang
- School of Pharmacy, Hunan Provincial Maternal and Child Health Care Hospital, University of South China, Hengyang, 421001, China
| | - Zhishuo Miao
- Key Laboratory of Drug Metabolism Research and Evaluation of the State Drug Administration, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Yinhuang Gao
- Key Laboratory of Drug Metabolism Research and Evaluation of the State Drug Administration, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - ZhiZhong Xie
- School of Pharmacy, Hunan Provincial Maternal and Child Health Care Hospital, University of South China, Hengyang, 421001, China
| | - Menghua Liu
- Key Laboratory of Drug Metabolism Research and Evaluation of the State Drug Administration, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China.
| | - Wei Zou
- School of Pharmacy, Hunan Provincial Maternal and Child Health Care Hospital, University of South China, Hengyang, 421001, China.
| |
Collapse
|
13
|
Cai J, Zhou H, Liu M, Zhang D, Lv J, Xue H, Zhou H, Zhang W. Host immunity and intracellular bacteria evasion mechanisms: Enhancing host-directed therapies with drug delivery systems. Int J Antimicrob Agents 2025; 65:107492. [PMID: 40107461 DOI: 10.1016/j.ijantimicag.2025.107492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 03/04/2025] [Accepted: 03/10/2025] [Indexed: 03/22/2025]
Abstract
Host-directed therapies (HDTs) have been investigated as a potential solution to combat intracellular and drug-resistant bacteria. HDTs stem from extensive research on the intricate interactions between the host and intracellular bacteria, leading to a treatment approach that relies on immunoregulation. To improve the bioavailability and safety of HDTs, researchers have utilized diverse drug delivery systems (DDS) to encapsulate and transport therapeutic agents to target cells. In this review, we first introduce the three mechanisms of bactericidal action and intracellular bacterial evasion: autophagy, reactive oxygen species (ROS), and inflammatory cytokines, with a particular focus on autophagy. Special attention is given to the detailed mechanism of xenophagy in clearing intracellular bacteria, a crucial selective autophagy process that specifically targets and degrades intracellular pathogens. Following this, we present the application of DDS to modulate these regulatory methods for intracellular bacteria elimination. By integrating insights from immunology and nanomedicine, this review highlights the emerging role of DDS in advancing HDTs for intracellular bacterial infections and paving the way for innovative therapeutic interventions.
Collapse
Affiliation(s)
- Jiayang Cai
- Department of Pharmaceutics, China Pharmaceutical University, Jiangsu, China
| | - Han Zhou
- Department of Pharmaceutics, China Pharmaceutical University, Jiangsu, China
| | - Mingwei Liu
- Department of Pharmaceutics, China Pharmaceutical University, Jiangsu, China
| | - Dingjian Zhang
- Department of Pharmaceutics, China Pharmaceutical University, Jiangsu, China
| | - Jingxuan Lv
- Department of Pharmaceutics, China Pharmaceutical University, Jiangsu, China
| | - Haokun Xue
- Department of Pharmaceutics, China Pharmaceutical University, Jiangsu, China
| | - Houcheng Zhou
- Department of Pharmaceutics, China Pharmaceutical University, Jiangsu, China
| | - Wenli Zhang
- Department of Pharmaceutics, China Pharmaceutical University, Jiangsu, China.
| |
Collapse
|
14
|
Wang Y, Mu H, Yang B, Yang C, Dong W, Wang J. USP7 - A novel target for controlling periodontal inflammation through modulation of macrophage polarization. Immunol Lett 2025; 273:106981. [PMID: 39946796 DOI: 10.1016/j.imlet.2025.106981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 01/11/2025] [Accepted: 02/09/2025] [Indexed: 03/12/2025]
Abstract
Disruption of local microbial irritation and host immune response can result in inflammation and tissue destruction in periodontitis. Studies on the modulation of macrophage polarization could help attenuate immune responses in periodontal tissues. To investigate the effect of ubiquitin-specific protease-7 (USP7) and its inhibitor P5091 on the polarization of macrophages in periodontitis, gene expression in periodontitis tissues and normal control were analyzed via single-cell RNA sequencing data and mice model experimental periodontitis. RAW264.7 cells were induced to M1 polarization with LPS + IFN-γ and M2 polarization with IL-4. USP7 was knocked down using lentivirus, and the effect of USP7 inhibitor P5091 on macrophage polarization was comparatively analyzed. The expression of Usp7 and polarization markers were detected by qRT-PCR. Western blot was used to examine the polarization markers and pathway-associated proteins. Results indicated that USP7 expression was elevated in tissues affected by periodontitis. Periodontitis macrophages and M1 polarized macrophages had higher USP7 expression. Knockdown of USP7 revealed an inhibition of both M1 and M2 macrophage polarization. Inhibition of USP7 with P5091 resulted in the decreased expression of M1 polarization markers and phosphorylation of P65, but the increased expression of M2 polarization markers and phosphorylation of STAT6. In conclusion, USP7 is involved in regulating macrophage polarization in periodontitis and its inhibitor P5091 may contribute to the prevention of periodontitis.
Collapse
Affiliation(s)
- Yan Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, 430079, China
| | - Hailin Mu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, 430079, China
| | - Baochen Yang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, 430079, China
| | - Chang Yang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, 430079, China
| | - Wei Dong
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, 430079, China
| | - Jiawei Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, 430079, China.
| |
Collapse
|
15
|
Zhou SL, Zhong LL, Wu YL, Ji SW, Li Y, Niu N. The role of ion channels in the regulation of dendritic cell function. Cell Calcium 2025; 128:103031. [PMID: 40253771 DOI: 10.1016/j.ceca.2025.103031] [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: 09/19/2024] [Revised: 04/12/2025] [Accepted: 04/16/2025] [Indexed: 04/22/2025]
Abstract
Ion channels, membrane proteins that facilitate the transport of various inorganic ions across hydrophobic cellular lipid membranes, are ubiquitous in a wide variety of cell and tissue types. They are involved in establishing the cell membrane potential and play a role in various physiological activities by regulating ion concentrations within the cell. Dendritic cells (DCs) are specialised antigen-presenting cells found mainly on the surface of the body (skin and mucous membranes), in the mesenchyme of most organs, in the T-cell compartment of the spleen and in lymph nodes. DCs exert an important influence on the regulation of inflammation by activating T cells and producing cytokines. Studies have shown that ion channels expressed in DCs contribute to the regulation of the immune response, making them a key component of the immune system. This review summarises the major scientific advances in understanding the functional impact of ion channels (calcium channels, sodium channels and aquaporin) in DCs, including the regulation of inflammatory responses, antigen presentation, maturation, migration and cytokine production, to inform ongoing studies of ion channel function in DCs.
Collapse
Affiliation(s)
- Shi-Li Zhou
- School of Pharmacy, North Sichuan Medical College, Nanchong, 637000, China
| | - Lan-Lan Zhong
- School of Pharmacy, North Sichuan Medical College, Nanchong, 637000, China
| | - Yi-Lan Wu
- School of Pharmacy, North Sichuan Medical College, Nanchong, 637000, China
| | - Si-Wen Ji
- Office of Academic Affairs, North Sichuan Medical College, Nanchong, 637000, China
| | - Yong Li
- School of Pharmacy, North Sichuan Medical College, Nanchong, 637000, China
| | - Na Niu
- School of Pharmacy, North Sichuan Medical College, Nanchong, 637000, China..
| |
Collapse
|
16
|
Jordan PM, Peltner LK, Bachmann V, Dahlke P, Pace S, Thost L, Hörcher LC, Nischang V, Temml V, Rossi A, Werz O. Uncovering anti-inflammatory natural products that synergize with supplemented omega-3 PUFA for eliciting endogenous inflammation resolution signals. Biomed Pharmacother 2025; 188:118190. [PMID: 40449167 DOI: 10.1016/j.biopha.2025.118190] [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: 03/18/2025] [Revised: 05/13/2025] [Accepted: 05/21/2025] [Indexed: 06/03/2025] Open
Abstract
Inflammation resolution is governed by specialized pro-resolving mediators (SPM), mainly formed from omega-3 polyunsaturated fatty acids (n-3 PUFA) by lipoxygenases (LOX), which terminate inflammatory processes and facilitate healing and tissue repair. Promoting endogenous SPM formation, besides therapeutic SPM application, is an innovative concept for intervention in inflammatory diseases, achievable by allosteric 15-LOX activation. Here, targeted screening of the 29 most frequently applied anti-inflammatory natural products using lipid mediator metabololipidomics uncovered the acylphloroglucinols hyperforin, arzanol, garcinol, Myrtucommulone A and the lignan magnolol as potent 15-LOX activators to elicit robust SPM production in resting human M2-like macrophages. Simultaneous n-3 PUFA supplementation synergistically enhanced SPM formation in these M2-like macrophages, most strikingly with magnolol. Comprehensive targeted metabololipidomics in activated human polymorphonuclear leukocytes, monocytes, and M1-/M2-like macrophages revealed shifts from pro-inflammatory cyclooxygenase (COX) and 5-LOX products to pro-resolving 15-LOX products by magnolol and acylphloroglucinols. Finally, using zymosan-induced peritonitis in mice, application of a magnolol/n-3 PUFA combination confirmed synergistic SPM elevation in vivo. Together, we established an approach based on synergism of natural allosteric 15-LOX activators and supplemented n-3 PUFA to accomplish SPM-based resolution pharmacology.
Collapse
Affiliation(s)
- Paul M Jordan
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Philosophenweg 14, Jena 07743, Germany.
| | - Lukas K Peltner
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Philosophenweg 14, Jena 07743, Germany
| | - Vivien Bachmann
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Philosophenweg 14, Jena 07743, Germany
| | - Philipp Dahlke
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Philosophenweg 14, Jena 07743, Germany
| | - Simona Pace
- Department for the Promotion of Human Science and Quality of Life, Chair of Pharmacology, "San Raffaele Open University of Rome", Rome 00163, Italy; Department of Pharmacy, University of Salerno, Via G. Paolo II, Fisciano 84084, Italy
| | - Lorenz Thost
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Philosophenweg 14, Jena 07743, Germany
| | - Lea Celine Hörcher
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Philosophenweg 14, Jena 07743, Germany
| | - Vivien Nischang
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Philosophenweg 14, Jena 07743, Germany
| | - Veronika Temml
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy, Paracelsus Medical University, Strubergasse 21, Salzburg 5020, Austria
| | - Antonietta Rossi
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples I-80131, Italy
| | - Oliver Werz
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Philosophenweg 14, Jena 07743, Germany.
| |
Collapse
|
17
|
Tan Q, Cao X, Zou F, Wang H, Xiong L, Deng S. Spatial Heterogeneity of Intratumoral Microbiota: A New Frontier in Cancer Immunotherapy Resistance. Biomedicines 2025; 13:1261. [PMID: 40427087 PMCID: PMC12108975 DOI: 10.3390/biomedicines13051261] [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: 04/28/2025] [Revised: 05/18/2025] [Accepted: 05/20/2025] [Indexed: 05/29/2025] Open
Abstract
The intratumoral microbiota, as an important component of the tumor microenvironment, is increasingly recognized as a key factor in regulating responses to cancer immunotherapy. Recent studies have revealed that the intratumoral microbiota is not uniformly distributed but instead exhibits significant spatial heterogeneity, with its distribution patterns influenced by factors such as tumor anatomy, local immune status, and therapeutic interventions. This spatial heterogeneity not only alters the interactions between microbes and the host immune system but may also reshape the immunogenic and immunosuppressive landscapes of tumors. The enrichment or depletion of microbiota in different tumor regions can influence immune cell infiltration patterns, metabolic pathway activities, and immune checkpoint molecule expression, thereby driving the development of resistance to immunotherapy. Moreover, certain bacterial metabolites form concentration gradients between the tumor core and margins, thereby regulating immune cell function. Therefore, understanding and manipulating the spatial distribution of intratumoral microbiota, particularly in resistant patients, holds promise for developing new strategies to overcome immunotherapy resistance. In the future, precise modulation strategies targeting microbial spatial heterogeneity, such as engineered bacterial vectors, probiotic combinations, and phage therapy, may open new avenues for immunotherapy.
Collapse
Affiliation(s)
- Qiwen Tan
- Department of Infectious Disease, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China;
| | - Xiongjing Cao
- Department of Nosocomial Infection Management, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China;
| | - Falong Zou
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (F.Z.); (H.W.)
| | - Hanwenchen Wang
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (F.Z.); (H.W.)
| | - Lijuan Xiong
- Department of Infectious Disease, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China;
- Department of Nosocomial Infection Management, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China;
| | - Shenghe Deng
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (F.Z.); (H.W.)
- Center for Liver Transplantation, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
18
|
Zhu X, Li Q, Wu J, Ju Z. Discovery of Safe COX-2 Inhibitors: Achieving Reduced Colitis Side Effects through Balanced COX Inhibition. ChemMedChem 2025; 20:e202500096. [PMID: 40012482 DOI: 10.1002/cmdc.202500096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 02/21/2025] [Accepted: 02/25/2025] [Indexed: 02/28/2025]
Abstract
The severe adverse effects associated with imbalanced cyclooxygenase-2 (COX-2) inhibition continue to pose significant challenges in the development of contemporary anti-inflammatory drugs. In recent years, the approach to COX-2 inhibitor drug development has shifted from a focus on highly selective inhibition of COX-2 to a strategy that emphasizes more moderate selectivity. The amino acid sequence and structural similarities between inducible COX-2 and constitutive cyclooxygenase-1 (COX-1) isoforms present both substantial opportunities and challenges for the design of next generation of balanced COX-2 inhibitors. As part of our ongoing research into the discovering novel and safer COX-2 inhibitors, we reported herein a highly potent and balanced COX-2 inhibitor 21 d (IC50 value=1.35 μM, selectivity profile (IC50 (COX-1)/IC50 (COX-2)=22.34)). In vivo assays demonstrated that 21 d significantly alleviated histological damage and provided robust protection against dextran sulfate sodium (DSS)-induced acute colitis.
Collapse
Affiliation(s)
- Xinlin Zhu
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Qin Li
- Chongqing Traditional Chinese Medicine Hospital, Chongqing, 400000, China
| | - Junhui Wu
- College of Chemical Engineering, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Zhiran Ju
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, 310014, China
| |
Collapse
|
19
|
Bi Y, Xu Y, Li T, Yuan T. Anti-Inflammatory Furylidene Tetronic Acid Derivatives from an Endophytic Fungus Hypoxylon monticulosum ZZ14. JOURNAL OF NATURAL PRODUCTS 2025. [PMID: 40378411 DOI: 10.1021/acs.jnatprod.5c00291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2025]
Abstract
Naturally occurring furylidene tetronic acids were rare and structurally unique, typically with a furylidene connected to a furan-2,4-(3H, 5H)dione (tetronic acid moiety). In the current study, seven furylidene tetronic acid derivatives (1-7) were isolated and identified from the endophytic fungus Hypoxylon monticulosum ZZ14, including three new ones, hypoxytetronic acids A and B (1 and 2) and 2',3'-dihydronodulisporacid A methyl ester (3). Compounds 1 and 2 possess an unusual skeleton, with two tetrahydrofurans attached to the tetronic acid moiety. Their absolute configurations were elucidated by custom-DP4+ probability analysis of NMR chemical shifts and ECD calculations. Nodulisporacid A methyl ester (7) significantly inhibited the production of interleukin 6 (IL-6), interleukin 10 (IL-10), nitric oxide (NO), and inducible nitric oxide synthase (iNOS), comparable to the positive control dexamethasone. Several analogues of nodulisporacid A methyl ester were synthesized, and structure-activity relationship studies were performed, suggesting that the conjugated fragment of 1/2/3/1'/2'/3' and the methyl ester group may be the pharmacophores.
Collapse
Affiliation(s)
- Yufang Bi
- Jiangxi Province Key Laboratory of Natural and Biomimetic Drugs Research, School of Health, Jiangxi Normal University, Nanchang 330022, China
- College of Chemistry and Material, Jiangxi Normal University, Nanchang 330022, China
| | - Yuelun Xu
- College of Chemistry and Material, Jiangxi Normal University, Nanchang 330022, China
| | - Tianzhi Li
- Jiangxi Province Key Laboratory of Natural and Biomimetic Drugs Research, School of Health, Jiangxi Normal University, Nanchang 330022, China
| | - Tao Yuan
- Jiangxi Province Key Laboratory of Natural and Biomimetic Drugs Research, School of Health, Jiangxi Normal University, Nanchang 330022, China
| |
Collapse
|
20
|
Gu D, Hu L, Yang K, Yuan W, Shan D, Gao J, Li J, Gimple RC, Dixit D, Zhu Z, Li D, Wu Q, Shi Z, Wang Y, Zhao N, Yang K, Shao J, Lin F, Wang Q, Jin G, Chen Y, Qian X, Hu Z, Li C, Zhang N, You Y, Liu J, Zhang Q, Zhang J, Rich JN, Wang X. Stress-induced pro-inflammatory glioblastoma stem cells secrete TNFAIP6 to enhance tumor growth and induce suppressive macrophages. Dev Cell 2025:S1534-5807(25)00287-4. [PMID: 40403724 DOI: 10.1016/j.devcel.2025.04.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 01/21/2025] [Accepted: 04/30/2025] [Indexed: 05/24/2025]
Abstract
Glioblastoma (GBM) is the most aggressive primary intracranial tumor, with glioblastoma stem cells (GSCs) enforcing the intratumoral hierarchy. The inflammatory microenvironment influences tumor development at varying stages, while the underlying mechanism of GSCs facing pro-inflammatory stress remains unclear. Here, we show that, in human GBM, pro-inflammatory stress from pro-inflammatory macrophages (pTAMs) maintains GSC proliferation and self-renewal. Tumor necrosis factor alpha-induced protein 6 (TNFAIP6), as a responder in patient-derived GSCs to pro-inflammatory stress tumor necrosis factor alpha (TNF-α) from human pTAMs, promotes tumor growth through binding epidermal growth factor (EGF) and prolonging EGF receptor (EGFR)-phosphatidylinositol 3-kinase (PI3K)-protein kinase B (AKT) signaling activation. Meanwhile, pro-inflammatory stress-induced patient-derived GSCs secrete TNFAIP6 to transform macrophage phenotype from pTAMs to inflammatory-suppressive macrophages (sTAMs). Collectively, pharmacological or genetic disruption of TNFAIP6 autocrine and paracrine communication between patient-derived GSCs and TAMs inhibited GSC proliferation and self-renewal in vitro and in patient-derived xenograft tumor-bearing mice, suggesting that TNFAIP6 is an effective target for GBM therapy.
Collapse
Affiliation(s)
- Danling Gu
- The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi 214000, Jiangsu, China; National Health Commission Key Laboratory of Antibody Techniques, Department of Cell Biology, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing 211166, Jiangsu, China; Institute for Brain Tumors, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing 210029, Jiangsu, China
| | - Lang Hu
- National Health Commission Key Laboratory of Antibody Techniques, Department of Cell Biology, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing 211166, Jiangsu, China; Institute for Brain Tumors, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing 210029, Jiangsu, China; Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China
| | - Kailin Yang
- Department of Radiation Oncology, Taussig Cancer Center, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Wei Yuan
- Department of Pathology, The Yancheng Clinical College of Xuzhou Medical University, The First People's Hospital of Yancheng, Yancheng 224005, Jiangsu, China
| | - Danyang Shan
- National Health Commission Key Laboratory of Antibody Techniques, Department of Cell Biology, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing 211166, Jiangsu, China; Institute for Brain Tumors, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing 210029, Jiangsu, China
| | - Jiancheng Gao
- National Health Commission Key Laboratory of Antibody Techniques, Department of Cell Biology, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing 211166, Jiangsu, China; Institute for Brain Tumors, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing 210029, Jiangsu, China
| | - Jiahuang Li
- School of Biopharmacy, China Pharmaceutical University, Jiangsu 211198, China
| | - Ryan C Gimple
- Department of Medicine, Washington University School of Medicine, Washington University in St Louis, St. Louis, MO 63110, USA
| | - Deobrat Dixit
- Department of Neurology, University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh, PA 15213, USA
| | - Zhe Zhu
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Daqi Li
- National Health Commission Key Laboratory of Antibody Techniques, Department of Cell Biology, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing 211166, Jiangsu, China; Institute for Brain Tumors, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing 210029, Jiangsu, China; Department of Neurology, University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh, PA 15213, USA
| | - Qiulian Wu
- Department of Neurology, University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh, PA 15213, USA
| | - Zhumei Shi
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China
| | - Yingyi Wang
- Institute for Brain Tumors, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing 210029, Jiangsu, China; Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China
| | - Ningwei Zhao
- China Exposomics Institute, 781 Cai Lun Road, Shanghai 200120, China
| | - Kun Yang
- Department of Neurosurgery, Zhongda Hospital, Southeast University, Nanjing 210009, Jiangsu, China
| | - Junfei Shao
- The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi 214000, Jiangsu, China
| | - Fan Lin
- National Health Commission Key Laboratory of Antibody Techniques, Department of Cell Biology, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing 211166, Jiangsu, China
| | - Qianghu Wang
- Institute for Brain Tumors, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing 210029, Jiangsu, China
| | - Guangfu Jin
- Institute for Brain Tumors, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing 210029, Jiangsu, China
| | - Yun Chen
- The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi 214000, Jiangsu, China; Institute for Brain Tumors, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing 210029, Jiangsu, China
| | - Xu Qian
- Institute for Brain Tumors, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing 210029, Jiangsu, China
| | - Zhibin Hu
- Institute for Brain Tumors, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing 210029, Jiangsu, China
| | - Chaojun Li
- Institute for Brain Tumors, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing 210029, Jiangsu, China
| | - Nu Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Brain Function and Disease, Guangdong Translational Medicine Innovation Platform, Guangzhou 510080, Guangdong, China
| | - Yongping You
- Institute for Brain Tumors, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing 210029, Jiangsu, China; Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China
| | - Jian Liu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, China.
| | - Qian Zhang
- National Health Commission Key Laboratory of Antibody Techniques, Department of Cell Biology, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing 211166, Jiangsu, China; Institute for Brain Tumors, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing 210029, Jiangsu, China.
| | - Junxia Zhang
- Institute for Brain Tumors, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing 210029, Jiangsu, China; Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China.
| | - Jeremy N Rich
- Department of Neurology, University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh, PA 15213, USA.
| | - Xiuxing Wang
- The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi 214000, Jiangsu, China; National Health Commission Key Laboratory of Antibody Techniques, Department of Cell Biology, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing 211166, Jiangsu, China; Institute for Brain Tumors, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing 210029, Jiangsu, China; Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China; Jiangsu Cancer Hospital, Affiliated Cancer Hospital of Nanjing Medical University, Nanjing 210009, Jiangsu, China.
| |
Collapse
|
21
|
Ávila-Ortega A, Villa-de la Torre FE, Carrera-Figueiras C, Martínez-Rizo AB, Talavera-Pech WA, Torres-Romero JC, Ceballos-Góngora E, García-Martínez V, Pintor-Romero VG, Huchin-Chan C, Vilchis-Nestor AR, Arana-Argáez VE. Toxicological and anti-inflammatory activities of doxorubicin loaded onto pH-sensitive poly(β-amino ester) modified mesoporous silica nanoparticles in mice. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04244-2. [PMID: 40366401 DOI: 10.1007/s00210-025-04244-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 04/28/2025] [Indexed: 05/15/2025]
Abstract
Chemotherapy drugs used in inflammatory disorders and some cancers, while effective, often cause diverse side toxic effects, prompting research into alternatives to ameliorate damage in healthy tissues. One strategy is to encapsulate doxorubicin (DOX) chemotherapeutic agents into nano-vehicles, such as organic-functionalized mesoporous nanoparticles, to reduce systemic leakage and adverse effects, such as inflammation. This study analyzes the toxicological and anti-inflammatory effects of a drug-delivered system (DDS) based on MCM-41 mesoporous silica nanoparticles modified with a pH-sensitive poly(β-amino ester) named MCM-41-PbAE. The effects of both blank nanoparticles (MCM-41-PbAE) and those with encapsulated DOX (MCM-41-PbAE-DOX) were evaluated in LPS-stimulated mouse macrophages and in an in vivo inflammation model. Characterization of nanoparticles was carried out through TEM, DLS, FTIR Z-potential, and thermogravimetric tests. Macrophages were treated with MCM-41-PbAE and MCM-41-PbAE-DOX particles at several concentrations, and the production of proinflammatory and anti-inflammatory cytokines, nitric oxide (NO), and hydrogen peroxide (H2O2) levels were determined. Toxicological evaluation was performed in BALB/c mice and the in vivo anti-inflammatory effect was evaluated in a carrageenan-induced paw edema model and the measuring serum levels of pro-inflammatory cytokines. Results indicated that MCM-41-PbAE-DOX significantly reduces NO and H2O2 levels and pro-inflammatory cytokines (IL-1β, IL-6, and TNF-α) in dose-manner without affecting cell viability or causing toxicity in mice. Conversely, a reduction in carrageenan-induced paw edema and decreased levels of proinflammatory cytokines were noted, indicating the potential advantage of encapsulating DOX within MCM-41-PbAE nanoparticles, which could inhibit inflammation without compromising healthy cell viability.
Collapse
Affiliation(s)
- Alejandro Ávila-Ortega
- Cuerpo Académico de Química Fundamental y Aplicada, Facultad de Ingeniería Química, Universidad Autónoma de Yucatán, Mérida, Yucatán, México
| | - Fabiola Elizabeth Villa-de la Torre
- Laboratorio de Farmacología, Facultad de Química, Universidad Autónoma de Yucatán, Calle 43, No 613 X Calle 90, Col. Inalámbrica, CP. 97069, Mérida, Yucatán, México
| | - Cristian Carrera-Figueiras
- Cuerpo Académico de Química Fundamental y Aplicada, Facultad de Ingeniería Química, Universidad Autónoma de Yucatán, Mérida, Yucatán, México
| | - Abril Bernardette Martínez-Rizo
- Laboratorio de Farmacología, Facultad de Química, Universidad Autónoma de Yucatán, Calle 43, No 613 X Calle 90, Col. Inalámbrica, CP. 97069, Mérida, Yucatán, México
| | - William Alejandro Talavera-Pech
- Centro de Investigación en Corrosión, Universidad Autónoma de Campeche, Av. Héroe de Nacozari No. 480, San Francisco de Campeche, Campeche, 24079, México
| | - Julio César Torres-Romero
- Laboratorio de Bioquímica y Genética Molecular, Facultad de Química, Universidad Autónoma de Yucatán, Mérida, Yucatán, México
| | - Emanuel Ceballos-Góngora
- Laboratorio de Farmacología, Facultad de Química, Universidad Autónoma de Yucatán, Calle 43, No 613 X Calle 90, Col. Inalámbrica, CP. 97069, Mérida, Yucatán, México
| | - Valeria García-Martínez
- Laboratorio de Farmacología, Facultad de Química, Universidad Autónoma de Yucatán, Calle 43, No 613 X Calle 90, Col. Inalámbrica, CP. 97069, Mérida, Yucatán, México
| | - Valeria Guadalupe Pintor-Romero
- Laboratorio de Farmacología, Facultad de Química, Universidad Autónoma de Yucatán, Calle 43, No 613 X Calle 90, Col. Inalámbrica, CP. 97069, Mérida, Yucatán, México
| | - Claribel Huchin-Chan
- Laboratorio de Análisis Clínicos y de Servicio a La Comunidad, Facultad de Química, Universidad Autónoma de Yucatán, Mérida, Yucatán, México
| | | | - Víctor Ermilo Arana-Argáez
- Laboratorio de Farmacología, Facultad de Química, Universidad Autónoma de Yucatán, Calle 43, No 613 X Calle 90, Col. Inalámbrica, CP. 97069, Mérida, Yucatán, México.
| |
Collapse
|
22
|
Oliveira SDS, Honório da Silva JV, Vieira RDS, Moreira LFS, Bandeira PHA, Ramos BL, Silva MAA, Câmara NOS. SARM1: a key multifaceted component in immunoregulation, inflammation and neurodegeneration. Front Immunol 2025; 16:1521364. [PMID: 40433385 PMCID: PMC12106052 DOI: 10.3389/fimmu.2025.1521364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 04/22/2025] [Indexed: 05/29/2025] Open
Abstract
The downstream signaling pathways of TLR activation involve a family of adaptor proteins, including MYD88, TIRAP, TRIF, TRAM, and SARM1. The first four proteins stimulate inflammatory and antiviral responses, playing crucial roles in innate immunity against various pathogens. In contrast, SARM1 promotes immunity to microorganisms in invertebrate animals independently of TLRs, and negatively regulates inflammatory responses in metazoan organisms. SARM1 inhibits TRIF, reduces the activation of various inflammasomes, and induces mitochondrial damage and cell death to eliminate hyperactivated cells. This regulation is essential to ensure timely control of immune responses and to prevent excessive inflammation. Recently, it was discovered that SARM1 can hydrolyze NAD, a critical component of cellular metabolism. The reduction of NAD levels by SARM1 is linked to the progression of Wallerian degeneration following neuronal injury and may also play a role in the immunoregulation of lymphoid and myeloid cells. Since SARM1 can be pharmacologically modulated, it presents promising opportunities for developing treatments for inflammatory and neurodegenerative diseases.
Collapse
Affiliation(s)
- Samuel dos Santos Oliveira
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School – FMRP of the University of São Paulo – USP, Ribeirão Preto, Brazil
| | | | - Raquel de Souza Vieira
- Department of Immunology, Institute of Biomedical Science – ICB of the University of São Paulo – USP, São Paulo, Brazil
| | - Luís Felipe Serra Moreira
- Department of Immunology, Institute of Biomedical Science – ICB of the University of São Paulo – USP, São Paulo, Brazil
| | | | - Beatriz Leocata Ramos
- Department of Immunology, Institute of Biomedical Science – ICB of the University of São Paulo – USP, São Paulo, Brazil
| | - Marco Antônio Ataíde Silva
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School – FMRP of the University of São Paulo – USP, Ribeirão Preto, Brazil
| | - Niels Olsen Saraiva Câmara
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School – FMRP of the University of São Paulo – USP, Ribeirão Preto, Brazil
- Department of Immunology, Institute of Biomedical Science – ICB of the University of São Paulo – USP, São Paulo, Brazil
| |
Collapse
|
23
|
Wang S, Jaggi U, Oh JJ, Ghiasi H. IFNβ absence compensates for LAT functions in latency reactivation and T cell exhaustion. J Virol 2025:e0037425. [PMID: 40353667 DOI: 10.1128/jvi.00374-25] [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: 02/24/2025] [Accepted: 04/13/2025] [Indexed: 05/14/2025] Open
Abstract
Type I interferons (IFNs) generate strong antiviral immunity following viral infection in mice and humans. These type I IFNs are encoded by at least 14 IFNα genes and a single IFNβ gene. We showed that the absence of one of the IFNα genes (IFNα2A-/-) affected latency levels in herpes simplex virus type 1 (HSV-1) ocularly infected mice but not in infected control mice, and the absence of IFNα2A did not affect viral reactivation in ocularly infected mice. Since the role of IFNβ in HSV-1 latency reactivation and the potential effect of latency-associated transcript (LAT) on IFNβ activity is not known, we ocularly infected IFNβ-/- mice with different doses of LAT-plus [LAT(+)] and LAT-minus [LAT(-)] viruses. Wild-type (WT) control mice and IFNβ-/- mice were infected similarly. Virus titers in the eye, viral and cellular transcripts in the eye and trigeminal ganglia (TG) of infected mice on days 3 and 5 post-infection, eye disease, survival, latency-reactivation levels, and T cell exhaustion were measured in latently infected mice. Virus replication and viral and cellular transcripts in the eye of infected IFNβ-/- mice were similar to those in WT mice, while eye disease and survival in IFNβ-/- mice differed significantly from WT mice. WT mice infected with LAT(-) virus showed reduced latency, slower reactivation, and less T cell exhaustion than mice infected with LAT(+) virus. However, using different doses of each virus, latency levels, time of reactivation, and T cell exhaustion were similar between LAT(+) and LAT(-) viruses. These results suggest that the absence of IFNβ expression compensates for the function of LAT with regard to levels of latency, T cell exhaustion, and reactivation but does not affect viral and cellular transcripts during primary infection.IMPORTANCEInterferon β (IFNβ) is a type I interferon that plays an important role in controlling primary herpes simplex virus type 1 (HSV-1) infection. To evaluate the importance of IFNβ on HSV-1 latency reactivation and its relationship to LAT, we infected IFNβ-/- mice with LAT(+) and LAT(-) viruses. In the absence of IFNβ, latency levels in mice infected with LAT(-) virus were similar to those of mice infected with LAT(+) virus. The absence of IFNβ also reduced the time of reactivation in mice infected with LAT(-) virus to that of LAT(+) virus. Our results show a strong correlation between the functions of LAT and IFNβ during latent but not primary stages of HSV-1 infection.
Collapse
Affiliation(s)
- Shaohui Wang
- Center for Neurobiology and Vaccine Development, Ophthalmology Research, Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Ujjaldeep Jaggi
- Center for Neurobiology and Vaccine Development, Ophthalmology Research, Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Jay J Oh
- Center for Neurobiology and Vaccine Development, Ophthalmology Research, Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Homayon Ghiasi
- Center for Neurobiology and Vaccine Development, Ophthalmology Research, Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, California, USA
| |
Collapse
|
24
|
Villa de la Torre F, Tec Caamal EA, Rizo AM, Argáez RB, Pintor Romero VG, Yáñez-Barrientos E, Morales-Tirado DJ, Alonso-Castro AJ, Arana Argáez VE. In vivo and in vitro anti-inflammatory activity of the methanolic leaves extract of Gymnopodium floribundum Rolfe. JOURNAL OF ETHNOPHARMACOLOGY 2025; 347:119743. [PMID: 40204249 DOI: 10.1016/j.jep.2025.119743] [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: 10/28/2024] [Revised: 03/28/2025] [Accepted: 04/02/2025] [Indexed: 04/11/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Gymnopodium floribundum Rolfe, known locally as "Dzidzilche" or "Ts'its'ilche," is a native species from Mexico and Central America. In Mayan communities, this plant is used to relieve inflammation and diverse respiratory diseases such as colds, catarrh, bronchitis, and asthma. Usually, a decoction of leaves or flowers is prepared and administered orally. AIM This research explores the anti-inflammatory effects of the methanol extract of Gymnopodium floribundum Rolfe leaves (MGF) using in vitro and in vivo animal models of inflammation. METHODS MGF was characterized by GC-MS, and cytotoxicity was assessed using hemolysis and MTT assays. The antiphlogistic effect in vitro was measuring the release of cytokines, hydrogen peroxide, and nitric oxide in macrophages stimulated with LPS. Additionally, anti- and pro-inflammatory cytokines, prostaglandins, and leukotrienes in serum were quantified in carrageenan-induced mouse paw edema. Finally, 1-fluoro-2,4-dinitrobenzene (DNFB)-induced delayed-type hypersensitivity and TPA-induced ear edema models were analyzed. RESULTS Compounds found in MGF, such as D-pinitol and protocatechuic (3,4-dihydroxybenzoic) acid, are reported to exert anti-inflammatory effects. MGF showed no hemolytic or cytotoxic effects. Nevertheless, it displayed in vitro anti-inflammatory activity by decreasing the release of IL-6, IL-1β, TNF-α, hydrogen peroxide, and nitric oxide levels; on the other hand, it increased IL-10 production. Furthermore, the MGF significantly reduced inflammation in mouse models and reduced the release of leukotrienes, prostaglandins, and pro-inflammatory cytokines. CONCLUSION Gymnopodium floribundum Rolfe exhibits anti-inflammatory activity by suppressing pro-inflammatory mediators, altering cell migration mechanisms, and raising IL-10 production.
Collapse
Affiliation(s)
- Fabiola Villa de la Torre
- Faculty of Chemistry, Laboratory of Pharmacology, Autonomous University of Yucatan, Yucatan, Mexico.
| | | | - Abril Martínez Rizo
- Faculty of Chemistry, Laboratory of Pharmacology, Autonomous University of Yucatan, Yucatan, Mexico.
| | - Rocío Borges Argáez
- Biotechnology Unit, Scientific Research Center of Yucatan (CICY), Yucatan, Mexico.
| | | | - Eunice Yáñez-Barrientos
- Division of Natural and Exact Sciences, Department of Chemistry, University of Guanajuato, Guanajuato, Mexico.
| | | | - Angel Josabad Alonso-Castro
- Division of Natural and Exact Sciences, Department of Chemistry, University of Guanajuato, Guanajuato, Mexico.
| | - Víctor E Arana Argáez
- Faculty of Chemistry, Laboratory of Pharmacology, Autonomous University of Yucatan, Yucatan, Mexico.
| |
Collapse
|
25
|
Bahmani F, Shayanmanesh M, Safari M, Alaei A, Yasaman Pouriafar, Rasti Z, Zaker F, Rostami S, Damerchiloo F, Safa M. Bone marrow microenvironment in myelodysplastic neoplasms: insights into pathogenesis, biomarkers, and therapeutic targets. Cancer Cell Int 2025; 25:175. [PMID: 40349084 PMCID: PMC12065391 DOI: 10.1186/s12935-025-03793-z] [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: 12/26/2024] [Accepted: 04/17/2025] [Indexed: 05/14/2025] Open
Abstract
Myelodysplastic neoplasms (MDS) represent a heterogeneous group of malignant hematopoietic stem and progenitor cell (HSPC) disorders characterized by cytopenia, ineffective hematopoiesis, as well as the potential to progress to acute myeloid leukemia (AML). The pathogenesis of MDS is influenced by intrinsic factors, such as genetic insults, and extrinsic factors, including altered bone marrow microenvironment (BMM) composition and architecture. BMM is reprogrammed in MDS, initially to prevent the development of the disease but eventually to provide a survival advantage to dysplastic cells. Recently, inflammation or age-related inflammation in the bone marrow has been identified as a key pathogenic mechanism for MDS. Inflammatory signals trigger stress hematopoiesis, causing HSPCs to emerge from quiescence and resulting in MDS development. A better understanding of the role of the BMM in the pathogenesis of MDS has opened up new avenues for improving diagnosis, prognosis, and treatment of the disease. This article provides a comprehensive review of the current knowledge regarding the significance of the BMM to MDS pathophysiology and highlights recent advances in developing innovative therapies.
Collapse
Affiliation(s)
- Forouzan Bahmani
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Maryam Shayanmanesh
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mahdi Safari
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Amirarsalan Alaei
- Immunology Research Center, Tabriz University of Medical Science, Tabriz, Iran
| | - Yasaman Pouriafar
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Rasti
- Department of Hematology, School of Allied Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Farhad Zaker
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Shahrbano Rostami
- Research Institute for Oncology, Hematology and Cell Therapy, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Damerchiloo
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Majid Safa
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
26
|
Hsu CY, Jasim SA, Rasool KH, H M, Kaur J, Jabir MS, Alhajlah S, Kumar A, Jawad SF, Husseen B. Divergent functions of TLRs in gastrointestinal (GI) cancer: Overview of their diagnostic, prognostic and therapeutic value. Semin Oncol 2025; 52:152344. [PMID: 40347779 DOI: 10.1016/j.seminoncol.2025.152344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2025] [Revised: 03/10/2025] [Accepted: 03/15/2025] [Indexed: 05/14/2025]
Abstract
The relationship between the innate immune signal and the start of the adaptive immune response is the central idea of this theory. By controlling the inflammatory and tissue-repair reactions to damage, the Toll-like receptors (TLRs), as a family of PRRs, have attracted increasing attention for its function in protecting the host against infection and preserving tissue homeostasis. Microbial infection, damage, inflammation, and tissue healing have all been linked to the development of malignancies, especially gastrointestinal (GI) cancers. Recently, increased studies on TLR recognition and binding, as well as their ligands, have significantly advanced our knowledge of the various TLR signaling pathways and offered therapy options for GI malignancies. Upon activation by pathogen-associated or damage-associated molecular patterns (DAMPs and PAMPs), TLRs trigger key pathways like NF-κB, MAPK, and IRF. NF-κB activation promotes inflammation, cell survival, and proliferation, often contributing to tumor growth, metastasis, and therapy resistance. MAPK pathways similarly drive uncontrolled cell growth and invasion, while IRF pathways modulate interferon production, yielding both anti-tumor and protumor effects. The resulting chronic inflammatory environment within tumors can foster progression, yet TLR activation can also stimulate beneficial anti-tumor immune responses. However, the functions of TLR expression in GI cancers and their diagnostic and prognostic along with therapeutic value have not yet entirely been elucidated. Understanding how TLR activation contributes to anti-cancer immunity against GI malignancies may hasten immunotherapy developments and increase patient survival.
Collapse
Affiliation(s)
- Chou-Yi Hsu
- Thunderbird School of Global Management, Arizona State University Tempe Campus, Phoenix, Arizona, USA
| | - Saade Abdalkareem Jasim
- Medical Laboratory Techniques Department, Al-maarif University College, Anbar, Iraq; Biotechnology Department, College of Applied Science, Fallujah University, Fallujah, Iraq
| | - Khetam Habeeb Rasool
- Department of Biology, College of Science, University of Mustansiriyah, Mustansiriyah, Iraq
| | - Malathi H
- Department of Biotechnology and Genetics, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Jaswinder Kaur
- Chandigarh Pharmacy College, Chandigarh Group of Colleges, Mohali, Punjab, India
| | - Majid S Jabir
- Department of Applied Sciences, University of Technology, Anbar, Iraq
| | - Sharif Alhajlah
- Department of Medical Laboratories, College of Applied Medical Sciences, Shaqra University, Shaqra, Saudi Arabia.
| | - Abhinav Kumar
- Department of Nuclear and Renewable Energy, Ural Federal University Named after the First President of Russia Boris Yeltsin, Ekaterinburg, Russia; Centre for Research Impact & Outcome, Chitkara University, Rajpura, Punjab, India; Institute of Engineering and Technology, Chitkara University, Rajpura, Punjab, India
| | - Sabrean F Jawad
- Department of Pharmacy, Al-Mustaqbal University College, Hillah, Babylon, Iraq
| | - Beneen Husseen
- Medical Laboratory Technique College, the Islamic University, Najaf, Iraq; Medical Laboratory Technique College, the Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq; Medical Laboratory Technique College, the Islamic University of Babylon, Babylon, Iraq
| |
Collapse
|
27
|
Alfirevic I, Cicak H, Saracevic A, Galic H, Simundic AM. Pentraxin 3 stability in serum and urine samples at -20 °C during a 4-week period. Lab Med 2025:lmae111. [PMID: 40339153 DOI: 10.1093/labmed/lmae111] [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] [Indexed: 05/10/2025] Open
Abstract
INTRODUCTION The aim of our study was to examine the stability of pentraxin 3 (PTX3) in serum and urine samples during 4-week storage at -20 °C. METHODS Ten routine leftover serum and urine samples were used in this study. The samples were stored at -20 °C over 4 weeks and analyzed each week in a single batch using the Human pentraxin 3 ELISA kit (BioVendor Group). Stability was expressed as a percentage difference between the initial measurement and the measurements taken weekly. Total error (33%) was used as the performance criterion for stability. RESULTS The concentration of PTX3 decreases with time during storage at -20 °C in both serum and urine. The percentage difference exceeded the set criterion in serum samples at the third week of storage (percentage difference = -23.5% [95% CI, -36.6% to -10.5%]) and at the first week of storage in urine samples (percentage difference = -41.5% [95% CI, -63.1% to ‒19.8%]). DISCUSSION Pentraxin 3 can be stored in serum samples for a maximum of 2 weeks at -20 °C before analysis to measure representative results. Because PTX3 is not stable in urine samples, such samples should be analyzed on the day of sample collection.
Collapse
Affiliation(s)
- Igor Alfirevic
- University Department of Surgery, University Hospital Sveti Duh, Zagreb, Croatia
- Faculty of Dental Medicine and Health, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia
- University North, Varazdin, Croatia
| | - Helena Cicak
- Department of Medical Laboratory Diagnostics, University Hospital Sveti Duh, Zagreb, Croatia
| | - Andrea Saracevic
- Department of Medical Laboratory Diagnostics, University Hospital Sveti Duh, Zagreb, Croatia
| | - Hrvoje Galic
- Department of Medical Laboratory Diagnostics, University Hospital Sveti Duh, Zagreb, Croatia
| | - Ana-Maria Simundic
- Department of Global Medical & Clinical Affairs, Greiner Bio-One, Kremsmünster, Austria
- University of Zagreb, Faculty of Pharmacy and Biochemistry, Zagreb, Croatia
| |
Collapse
|
28
|
Majdalawieh AF, Khatib BK, Terro TM. α-Mangostin Is a Xanthone Derivative from Mangosteen with Potent Immunomodulatory and Anti-Inflammatory Properties. Biomolecules 2025; 15:681. [PMID: 40427574 PMCID: PMC12108668 DOI: 10.3390/biom15050681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2025] [Revised: 04/28/2025] [Accepted: 05/06/2025] [Indexed: 05/29/2025] Open
Abstract
α-Mangostin, a bioactive xanthone derived from the Garcinia mangostana L. Clusiaceae (G. mangostana) fruit, has demonstrated significant anti-inflammatory and immunomodulatory properties. Chronic inflammation plays a critical role in the pathogenesis of various diseases, including metabolic disorders, autoimmune conditions, and cancer. Conventional anti-inflammatory therapies, such as non-steroidal anti-inflammatory drugs (NSAIDs), often carry undesirable side effects, prompting the need for safer, natural alternatives. This review consolidates the existing literature on the mechanisms by which α-mangostin exerts its anti-inflammatory effects, including the suppression of pro-inflammatory cytokines, modulation of immune cell activity, and inhibition of key signaling pathways such as nuclear factor-kappa B (NF-κB) and mitogen-activated protein kinase (MAPK). Additionally, α-mangostin exhibits immunomodulatory properties by influencing both innate and adaptive immune responses, affecting macrophage polarization, T cell differentiation, and cytokine production. Its efficacy has been observed in numerous disease models, including joint disorders, digestive and metabolic conditions, hepatic diseases, neurological disorders, and respiratory ailments. The potential therapeutic applications of α-mangostin as an anti-inflammatory agent warrant further investigation through preclinical and clinical studies to validate its efficacy and safety.
Collapse
Affiliation(s)
- Amin F. Majdalawieh
- Department of Biology, Chemistry and Environmental Sciences, College of Arts and Sciences, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates; (B.K.K.); (T.M.T.)
- Advanced Biosciences and Bioengineering Research Center, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates
| | - Bayan K. Khatib
- Department of Biology, Chemistry and Environmental Sciences, College of Arts and Sciences, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates; (B.K.K.); (T.M.T.)
| | - Tala M. Terro
- Department of Biology, Chemistry and Environmental Sciences, College of Arts and Sciences, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates; (B.K.K.); (T.M.T.)
| |
Collapse
|
29
|
Motta MA, Martin-Saldaña S, Beloqui A, Calderón M, Larrañaga A. Polypeptide-based multilayer capsules with anti-inflammatory properties: exploring different strategies to incorporate hydrophobic drugs. J Mater Chem B 2025; 13:5297-5314. [PMID: 40207430 DOI: 10.1039/d4tb01906g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/11/2025]
Abstract
More than 90% of drug candidates used in the drug development pipeline and about 40% of drugs on the market are poorly soluble in water based on the definition of the biopharmaceutical classification system. The advent of drug delivery approaches has represented a striking tool to overcome the challenges associated with the use of hydrophobic drugs, such as their low bioavailability and off-target effects. Drug carrier formulations composed of biodegradable and biocompatible polymers, such as polypeptides, have been explored as platforms to host poorly water-soluble drugs to prolong drug circulation, enhance their safety, reduce their immunogenicity, and promote their controlled release. In this work, we evaluated three strategies-co-precipitation, post-encapsulation, and conjugation-to incorporate a hydrophobic model drug, i.e., curcumin (CUR), into biodegradable multilayer capsules fabricated via a layer-by-layer (LbL) approach. Poly(L-lysine) (PLys) and poly(L-glutamic acid) (PGlu) were adopted as building blocks and alternately assembled onto calcium carbonate (CaCO3) microparticles to build a polypeptide-multilayer membrane, which acted as a barrier to control the release of the drug. The application of our three formulations in in vitro inflammatory models of THP-1 derived human macrophages and murine microglia showed a reduction of the inflammation with the suppression of three pivotal pro-inflammatory cytokines (i.e., interleukin (IL)-1β, IL-6, and tumor necrosis factor (TNF)-α). Moreover, the intracellular release of CUR detected upon uptake studies on activated microglia suggested that our systems could represent a potential therapeutic approach to reduce acute neuroinflammation and modulate microglia phenotype.
Collapse
Affiliation(s)
- Maria Angela Motta
- POLYMAT, Applied Chemistry Department, Faculty of Chemistry, University of the Basque Country UPV/EHU, Paseo Manuel de Lardizabal 3, 20018 Donostia-San Sebastián, Spain.
- Department of Mining-Metallurgy Engineering and Materials Science, POLYMAT, Bilbao School of Engineering, University of the Basque Country (UPV/EHU), Plaza Torres Quevedo 1, 48013 Bilbao, Spain.
| | - Sergio Martin-Saldaña
- POLYMAT, Applied Chemistry Department, Faculty of Chemistry, University of the Basque Country UPV/EHU, Paseo Manuel de Lardizabal 3, 20018 Donostia-San Sebastián, Spain.
| | - Ana Beloqui
- POLYMAT, Applied Chemistry Department, Faculty of Chemistry, University of the Basque Country UPV/EHU, Paseo Manuel de Lardizabal 3, 20018 Donostia-San Sebastián, Spain.
- IKERBASQUE, Basque Foundation for Science, Plaza Euskadi 5, 48009 Bilbao, Spain
| | - Marcelo Calderón
- POLYMAT, Applied Chemistry Department, Faculty of Chemistry, University of the Basque Country UPV/EHU, Paseo Manuel de Lardizabal 3, 20018 Donostia-San Sebastián, Spain.
- IKERBASQUE, Basque Foundation for Science, Plaza Euskadi 5, 48009 Bilbao, Spain
| | - Aitor Larrañaga
- Department of Mining-Metallurgy Engineering and Materials Science, POLYMAT, Bilbao School of Engineering, University of the Basque Country (UPV/EHU), Plaza Torres Quevedo 1, 48013 Bilbao, Spain.
| |
Collapse
|
30
|
Ma S, Ni J, Ye D, Kuang Y, Wang Z, Yang L. Human umbilical cord mesenchymal stem cells improve the survial of flaps by promoting angiogenesis in mice. Eur J Med Res 2025; 30:356. [PMID: 40312717 PMCID: PMC12046903 DOI: 10.1186/s40001-025-02602-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Accepted: 04/15/2025] [Indexed: 05/03/2025] Open
Abstract
BACKGROUND Flap necrosis post-operation disturbs surgeons during plastic and reconstructive surgery. This is caused by hypoperfusion and subsequent ischemia-reperfusion injury, where restricted blood flow followed by restored circulation paradoxically exacerbates tissue damage. Mesenchymal stem cells, which show multidirectional differentiation, provide hematopoietic support and are involved in immune regulation and anti-fibrosis, have inspired research on improving the blood supply of flaps. METHODS Primary human umbilical cord mesenchymal stem cells (HuMSCs), were obtained and subcultured for expansion. The cells of the third generation were incubated in a gelatin sponge. Thirty Kunming mice were randomly divided into three groups, and saline, HuMSCs, and HuMSCs-CM were injected preoperatively into the skin of the back. The vessel density was assessed on the tenth day. Forty-eight Kunming mice were divided into two groups. Group A was subdivided into the saline group, HuMSCs, and HuMSCs-CM groups and pretreated as described above. In Group B, the intervention was changed from injection to subcutaneous embedding. Random flaps were made on the back in both groups on the tenth day after pretreatment. The survival rate of the flap was calculated on the seventh day. RESULTS HuMSCs-CM significantly increased the microvessel density on the tenth day after pretreatment. The flap survival rate was higher in the cell and CM groups, rising from approximately 13% to 60% in Group A, and to about 75% in Group B. Moreover, subcutaneous embedding of cell-carrying gelatin sponges improved flap survival compared to other interventions. CONCLUSION Improved cell incubation conditions can enhance its utility. The application of HuMSCs and their conditioned medium promoted the survival of the flap by inducing neovasculogenesis.
Collapse
Affiliation(s)
- Siyi Ma
- Department of Plastic Surgery and Burns Center, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Jintao Ni
- Department of Plastic Surgery and Burns Center, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Danyan Ye
- Research Center for Translational Medicine, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Yuping Kuang
- Department of Plastic Surgery and Burns Center, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Zhixia Wang
- Department of Plastic Surgery and Burns Center, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Lujun Yang
- Department of Plastic Surgery and Burns Center, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China.
- Research Center for Translational Medicine, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China.
| |
Collapse
|
31
|
Gu S, Xu L, Huang B, Xiong K, Yang X, Ye J. Decoding Macrophage Dynamics: A Pathway to Understanding and Treating Inflammatory Skin Diseases. Int J Mol Sci 2025; 26:4287. [PMID: 40362523 PMCID: PMC12071885 DOI: 10.3390/ijms26094287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2025] [Revised: 04/27/2025] [Accepted: 04/29/2025] [Indexed: 05/15/2025] Open
Abstract
Psoriasis and atopic dermatitis (AD) are both chronic inflammatory skin diseases. Their pathogenesis remains incompletely understood. The polarization states of macrophages, as a crucial part of the innate immune system, are influenced by various factors such as cytokines, inflammatory mediators, and epigenetics. Research has demonstrated that macrophages play a "double-edged sword" role in the pathological process of inflammatory skin diseases: they both drive inflammation progression and participate in tissue repair. This article summarizes the roles of macrophages in the inflammatory development and tissue homeostasis of psoriasis and atopic dermatitis. It explores the impact of different factors on macrophages and inflammatory skin diseases. In conclusion, understanding the classification and plasticity of macrophages is crucial for a deeper understanding of the pathogenesis of psoriasis and AD and the development of personalized treatments.
Collapse
Affiliation(s)
- Shengliang Gu
- The First School of Clinical Medicine, Yunnan University of Chinese Medicine, Kunming 650500, China; (S.G.); (L.X.); (B.H.)
- Yunnan Provincial Clinical Medical Centre for Traditional Chinese Medicine Project (Dermatology), Kunming 650500, China
| | - Lei Xu
- The First School of Clinical Medicine, Yunnan University of Chinese Medicine, Kunming 650500, China; (S.G.); (L.X.); (B.H.)
| | - Bin Huang
- The First School of Clinical Medicine, Yunnan University of Chinese Medicine, Kunming 650500, China; (S.G.); (L.X.); (B.H.)
| | - Kai Xiong
- The First School of Clinical Medicine, Guizhou University of Chinese Medicine, Guiyang 550025, China;
| | - Xuesong Yang
- The First School of Clinical Medicine, Yunnan University of Chinese Medicine, Kunming 650500, China; (S.G.); (L.X.); (B.H.)
- Yunnan Provincial Clinical Medical Centre for Traditional Chinese Medicine Project (Dermatology), Kunming 650500, China
| | - Jianzhou Ye
- The First School of Clinical Medicine, Yunnan University of Chinese Medicine, Kunming 650500, China; (S.G.); (L.X.); (B.H.)
- Yunnan Provincial Clinical Medical Centre for Traditional Chinese Medicine Project (Dermatology), Kunming 650500, China
| |
Collapse
|
32
|
Qian S, Wang X, Guo Y, He W, Yang J, Chen H, Li R, Su L, Wang X, Shao Y, Wang B. Synchronous Sterilization and Immunoreaction Termination for Corneal Transparency Protection in Treating Pseudomonas aeruginosa Induced Bacterial Keratitis. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2419209. [PMID: 40166821 DOI: 10.1002/adma.202419209] [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: 12/07/2024] [Revised: 03/24/2025] [Indexed: 04/02/2025]
Abstract
In the treatment of infectious keratitis, therapeutic strategies often prioritize enhancing bactericidal efficacy. However, endotoxins released from Gram-negative bacteria cause inflammatory reaction, leading to corneal structural damage and scar formation. Given that polymyxin B (PMB) can bind and neutralize lipopolysaccharide (LPS), this study employs large-pore mesoporous silica nanoparticles (lMSNs) grafted with PMB as carriers for cationic antibacterial carbon quantum dots (CQDs) to prepare CQD@lMSN-PMB, which enables synchronous sterilization and endotoxin neutralization. In the acidic infectious microenvironment, the accelerated release of CQDs eliminates 99.88% bacteria within 2 h, effectively substituting immune mediated sterilization. Notably, CQD@lMSN-PMB exhibits exceptional LPS neutralization performance (2.22 µg LPS/mg CQD@lMSN-PMB) due to its high specific surface area. In an infectious keratitis model, inflammation subsides significantly within the first day of CQD@lMSN-PMB intervention and is completely resolved by day 3. By day 2, interleukin-1β, interleukin-6 and tumor necrosis factor-α in CQD@lMSN-PMB group decrease by 86.99%, 91.15%, and 77.56%, respectively, compared to the CQDs-only sterilization group. Ultimately, corneal integrity and transparency are preserved, with suppressed expressions of fibrosis-related factors including matrix metalloproteinase 9, transforming growth factor-β and α-smooth muscle actin. Therefore, this synchronous sterilization and endotoxin neutralization strategy outperforms monotherapy strategies focused solely on sterilization or endotoxin neutralization.
Collapse
Affiliation(s)
- Siyuan Qian
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325000, China
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Xuan Wang
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325000, China
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
- NMPA Key Laboratory for Clinical Research and Evaluation of Medical Devices and Drug for Ophthalmic Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Yishun Guo
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325000, China
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Wenfang He
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
- NMPA Key Laboratory for Clinical Research and Evaluation of Medical Devices and Drug for Ophthalmic Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Jianhua Yang
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
- NMPA Key Laboratory for Clinical Research and Evaluation of Medical Devices and Drug for Ophthalmic Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Hao Chen
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325000, China
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Renlong Li
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
- NMPA Key Laboratory for Clinical Research and Evaluation of Medical Devices and Drug for Ophthalmic Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Lili Su
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
- NMPA Key Laboratory for Clinical Research and Evaluation of Medical Devices and Drug for Ophthalmic Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Xinyi Wang
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325000, China
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
- NMPA Key Laboratory for Clinical Research and Evaluation of Medical Devices and Drug for Ophthalmic Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Yi Shao
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Eye Diseases, Shanghai, 200080, China
| | - Bailiang Wang
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325000, China
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
- NMPA Key Laboratory for Clinical Research and Evaluation of Medical Devices and Drug for Ophthalmic Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| |
Collapse
|
33
|
Pei J, Chen S, Ke Q, Pang A, Niu M, Li N, Li J, Wang Z, Wu H, Nie P. Immune response to polystyrene microplastics: Regulation of inflammatory response via the ROS-driven NF-κB pathway in zebrafish (Danio rerio). AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2025; 282:107308. [PMID: 40058300 DOI: 10.1016/j.aquatox.2025.107308] [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: 01/11/2025] [Revised: 02/16/2025] [Accepted: 03/02/2025] [Indexed: 04/05/2025]
Abstract
There is increasing apprehension regarding the rising prevalence of microplastics (MPs) in aquatic ecosystems. Although MPs cause toxicological effect on fish via diverse pathways, the precise immunotoxicological mechanism is yet to be fully understood. Utilizing zebrafish in early developmental stages and zebrafish embryonic fibroblast (ZF4) as models, this study delved into the immune response elicited by polystyrene MPs (PS-MPs). It was observed that larvae predominantly accumulate 3 μm PS-MPs in their intestines through ingestion, leading to notable changes in locomotor behavior and histopathological alterations. Further investigation revealed that short-term exposure to PS-MPs triggers oxidative stress (OS) and inflammation in zebrafish. This is evidenced by the upregulation of OS and inflammation-related genes, increased levels of reactive oxygen species (ROS), malonaldehyde (MDA), and inflammatory cytokines, altered activities of antioxidant enzymes, along with induced recruitment of leukocyte in larvae. Cellular assays confirmed that PS-MPs elevate intracellular ROS in ZF4 cells and enhance the nuclear translocation of NF-κB P65. Notably, the activation of NF-κB and the upsurge in inflammatory cytokines can be mitigated by inhibiting ROS. This research highlights the significance of the ROS-triggered NF-κB signaling cascade in PS-MPs-mediated inflammation within zebrafish, illuminating the possible processes that underlie the innate immune system of fish toxicity caused by MPs.
Collapse
Affiliation(s)
- Jincheng Pei
- Hubei Key Laboratory of Multi-media Pollution Cooperative Control in Yangtze Basin, School of Environmental Science & Engineering, Huazhong University of Science and Technology (HUST), Wuhan, Hubei Province 430074, PR China
| | - Shannan Chen
- State Key Laboratory of Freshwater Ecology and Biotechnology and Key Laboratory of Aquaculture Disease Control, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei Province 430072, PR China
| | - Qingxia Ke
- Yangxin County Fishery Service Center, Huangshi, Hubei Province 435200, PR China
| | - Anning Pang
- State Key Laboratory of Freshwater Ecology and Biotechnology and Key Laboratory of Aquaculture Disease Control, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei Province 430072, PR China
| | - Mengmeng Niu
- State Key Laboratory of Freshwater Ecology and Biotechnology and Key Laboratory of Aquaculture Disease Control, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei Province 430072, PR China
| | - Nan Li
- State Key Laboratory of Freshwater Ecology and Biotechnology and Key Laboratory of Aquaculture Disease Control, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei Province 430072, PR China
| | - Jiayi Li
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, Shandong Province 266109, PR China
| | - Zhi Wang
- Yangxin County Fishery Service Center, Huangshi, Hubei Province 435200, PR China
| | - Hongjuan Wu
- Hubei Key Laboratory of Multi-media Pollution Cooperative Control in Yangtze Basin, School of Environmental Science & Engineering, Huazhong University of Science and Technology (HUST), Wuhan, Hubei Province 430074, PR China.
| | - Pin Nie
- State Key Laboratory of Freshwater Ecology and Biotechnology and Key Laboratory of Aquaculture Disease Control, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei Province 430072, PR China; School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, Shandong Province 266109, PR China.
| |
Collapse
|
34
|
Markovic MD, Panic VV, Pjanovic RV. Polymeric Nanosystems: A Breakthrough Approach to Treating Inflammation and Inflammation Related Diseases. Biopolymers 2025; 116:e70012. [PMID: 40104970 DOI: 10.1002/bip.70012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 02/17/2025] [Accepted: 03/02/2025] [Indexed: 03/20/2025]
Abstract
Inflammation processes can cause mild to severe damage in the human body and can lead to a large number of inflammation-related diseases (IRD) such as cancer, neural, vascular, and pulmonary diseases. Limitations of anti-inflammatory drugs (AID) application are reflected in high therapeutic doses, toxicity, low bioavailability and solubility, side effects, etc. Polymeric nanosystems (PS) have been recognized as a safe and effective technology that is able to overcome these limitations by AID encapsulation and is able to answer to the specific demands of the IRD treatment. PS are attracting great attention due to their versatility, biocompatibility, low toxicity, fine-tuned properties, functionality, and ability for precise delivery of anti-inflammatory drugs to the targeted sites in the human body. This article offers an overview of three classes of polymeric nanosystems: a) dendrimers, b) polymeric micelles and polymeric nanoparticles, and c) polymeric filomicelles, as well as their properties, preparation, and application in IRD treatment. In the future, the number of PS formulations in clinical practice will certainly increase.
Collapse
Affiliation(s)
- Maja D Markovic
- Innovation Center of Faculty of Technology and Metallurgy, University of Belgrade, Belgrade, Serbia
| | - Vesna V Panic
- Innovation Center of Faculty of Technology and Metallurgy, University of Belgrade, Belgrade, Serbia
| | - Rada V Pjanovic
- Faculty of Technology and Metallurgy, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
35
|
Yong H, Yun D, Xu F, Tang C, Chen D, Kan J, Huang J, Yu H, Liu J. Dialdehyde starch-epicatechin gallate conjugate alleviates inflammation in lipopolysaccharide-stimulated RAW264.7 cells and dextran sulfate sodium-induced colitis mice. Int J Biol Macromol 2025; 306:141343. [PMID: 39988158 DOI: 10.1016/j.ijbiomac.2025.141343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Revised: 02/09/2025] [Accepted: 02/19/2025] [Indexed: 02/25/2025]
Abstract
In this study, epicatechin gallate (ECG), a natural anti-inflammatory agent, was conjugated onto dialdehyde starch (DAS) to achieve high physiological stability. The anti-inflammatory effect of DAS-ECG conjugate was evaluated by lipopolysaccharide (LPS)-stimulated RAW264.7 cells and dextran sulfate sodium (DSS)-induced colitis mice models. Results showed that 25-800 μg/mL of DAS-ECG conjugate was non-cytotoxic to RAW264.7 cells. DAS-ECG conjugate effectively inhibited the abnormal morphology, the production of nitric oxide, tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), interleukin-1β (IL-1β) and reactive oxygen species, and the apoptosis of LPS-stimulated RAW264.7 cells in a dose-dependent manner. DAS-ECG conjugate significantly reduced the disease activity index, thymus atrophy, spleen enlargement, colon shortening and colon damage of DSS-induced colitis mice. Meanwhile, DAS-ECG conjugate remarkably reduced the levels of TNF-α, IL-6, IL-1β and malondialdehyde but increased the levels of superoxide dismutase and glutathione in the colon tissue of DSS-induced colitis mice. Moreover, DAS-ECG conjugate increased the relative abundance of beneficial bacteria (Akkermansia, Candidatus_Saccharimonas, unclassified_f_Muribaculaceae, Alistipes and Parabacteroides), promoted the production of short-chain fatty acids, and decreased the relative abundance of harmful bacterium (norank_f_Ruminococcaceae) in DSS-induced colitis mice. Therefore, DAS-ECG conjugate could be considered as a promising anti-inflammatory agent.
Collapse
Affiliation(s)
- Huimin Yong
- College of Food Science and Engineering, Yangzhou University, Yangzhou 225127, China
| | - Dawei Yun
- College of Food Science and Engineering, Yangzhou University, Yangzhou 225127, China
| | - Fengfeng Xu
- College of Food Science and Engineering, Yangzhou University, Yangzhou 225127, China
| | - Chao Tang
- College of Food Science and Engineering, Yangzhou University, Yangzhou 225127, China
| | - Dan Chen
- College of Food Science and Engineering, Yangzhou University, Yangzhou 225127, China
| | - Juan Kan
- College of Food Science and Engineering, Yangzhou University, Yangzhou 225127, China
| | - Jinbao Huang
- State Key Laboratory of Tea Plant Biology and Utilization, Anhui Agricultural University, Hefei 230036, China
| | - Hai Yu
- College of Food Science and Engineering, Yangzhou University, Yangzhou 225127, China
| | - Jun Liu
- College of Food Science and Engineering, Yangzhou University, Yangzhou 225127, China.
| |
Collapse
|
36
|
Sahu M, Paliwal T, Jain S, Verma K, Chakraborty D, Jaiswal S, Dwivedi J, Sharma S. Multifaceted Therapeutic Impacts of Cucurbitacin B: Recent Evidences From Preclinical Studies. Phytother Res 2025; 39:1966-1995. [PMID: 39963741 DOI: 10.1002/ptr.8454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 10/30/2024] [Accepted: 11/25/2024] [Indexed: 05/21/2025]
Abstract
The most prevalent and bioactive cucurbitacin is Cucurbitacin B (CuB, C32H46O8), which is a tetracyclic triterpene chiefly present in the Cucurbitaceae family. CuB has a wide spectrum of pharmacological properties namely antioxidant, anticancer, hepatoprotective, anti-inflammatory, antiviral, hypoglycaemic, insecticidal, and neuroprotective properties, owing to its ability to regulate several signaling pathways, including the Janus kinase/signal transducer and activator of transcription-3 (JAK/STAT3), AMP-activated protein kinase (AMPK), nuclear factor (NF)-κB, nuclear factor erythroid 2-related factor-2/antioxidant responsive element (Nrf2/ARE), phosphoinositide 3-kinase (PI3K)/Akt, mitogen-activated protein kinase (MAPK), Hippo-Yes-associated protein (YAP), focal adhesion kinase (FAK), cancerous inhibitor of protein phosphatase-2A/protein phosphatase-2A (CIP2A/PP2A), Wnt and Notch pathways. The present review highlights the medicinal attributes of Cucurbitacin B (CuB) with special emphasis on their signaling pathways to provide key evidence of its therapeutic utility in the near future.
Collapse
Affiliation(s)
- Meenal Sahu
- Department of Bioscience & Biotechnology, Banasthali Vidyapith, Banasthali, Rajasthan, India
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, Rajasthan, India
| | - Tripti Paliwal
- Department of Bioscience & Biotechnology, Banasthali Vidyapith, Banasthali, Rajasthan, India
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, Rajasthan, India
| | - Smita Jain
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Kishangarh, Rajasthan, India
| | - Kanika Verma
- Department of Internal Medicine, Division of Cardiology, LSU Health Sciences Center, Shreveport, Louisiana, USA
| | - Dipjyoti Chakraborty
- Department of Bioscience & Biotechnology, Banasthali Vidyapith, Banasthali, Rajasthan, India
| | - Shivangi Jaiswal
- Department of Chemistry, Banasthali Vidyapith, Banasthali, Rajasthan, India
| | - Jaya Dwivedi
- Department of Chemistry, Banasthali Vidyapith, Banasthali, Rajasthan, India
| | - Swapnil Sharma
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, Rajasthan, India
| |
Collapse
|
37
|
Rahmani NR, Jahanmard F, Hassani Najafabadi A, Flapper J, Dogan O, Khodaei A, Storm G, Croes M, Kruyt MC, Gawlitta D, Weinans H, Mastrobattista E, Amin Yavari S. Local delivery of lipid-based nanoparticles containing microbial nucleic acid for osteoimmunomodulation. Eur J Pharm Sci 2025; 208:107050. [PMID: 39988262 DOI: 10.1016/j.ejps.2025.107050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 01/27/2025] [Accepted: 02/21/2025] [Indexed: 02/25/2025]
Abstract
Osteoimmunomodulation is a strategy to promote bone regeneration in implants by modifying the immune environment. CpG-containing oligonucleotides type C (CpG ODN C) and Polyinosinic:polycytidylic acid (Poly[I:C]) are analogs of microbial nucleic acids that have been studied for various immunotherapeutic applications. This research investigates the potential of CpG ODN C and Poly(I:C) as an osteoimmunomodulatory agent for bone regenerative purposes. We encapsulated each nucleic acid in a lipid-based nanoparticle to facilitate the delivery into intracellular pathogen recognition receptors in immune cells. The lipid-based nanoparticles were ±250 nm in size with a negative charge (-36 to -40 mV) and an encapsulation efficiency of ±60 %. Lipid-based nanoparticles containing nucleic acids, Lip/CpG ODN C and Lip/Poly(I:C), increased the production of TNF, IL-6, and IL-10 by primary human macrophages compared to free-form nucleic acids. Conditioned medium from macrophages treated with CpG ODN C (10 µg/ml) and Lip/CpG ODN C (0.1, 1, and 10 µg/ml) promoted osteoblast differentiation of human mesenchymal stromal cells by 2.6-fold and 3-fold, respectively; no effect was seen for Lip/Poly(I:C). Bone implants were prepared, consisting of a biphasic calcium phosphate scaffold, bone morphogenetic protein (BMP) 2, and lipid-based nanoparticles suspended in gelatin methacryloyl (GelMA) hydrogel. Implants were evaluated for de novo bone formation in an extra-skeletal implantation model in rabbits for 5 weeks. Based on the particles suspended in GelMA, six groups of implants were prepared: Lip/CpG ODN C, Lip/Poly(I:C), Lip (empty), CpG ODN C, Poly(I:C), and a control group consisting of empty GelMA. After 5 weeks, healthy bone tissue formed in all of the implants with active osteoblast and osteoclast activity, however, the amount of new bone volume and scaffold degradation were similar for all implants. We suggest that the working concentrations of the nucleic acids employed were inadequate to induce a relevant inflammatory response. Additionally, the dosage of BMP-2 used may potentially mask the immune-stimulatory effect. Lip/CpG ODN C holds potential as a bioactive agent for osteoimmunomodulation, although further in vivo demonstration should corroborate the current in vitro findings.
Collapse
Affiliation(s)
- N R Rahmani
- Department of Orthopedics, University Medical Center Utrecht, Heidelberglaan 100, GA 3508, Utrecht, the Netherlands; Regenerative Medicine Center Utrecht, University Utrecht, Heidelberglaan 8, CS 3584, Utrecht, the Netherlands.
| | - F Jahanmard
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, CG 3584, Utrecht, the Netherlands.
| | - A Hassani Najafabadi
- Terasaki Institute for Biomedical Innovation, 21100 Erwin St., Woodland Hills, 91367, Los Angeles, United States.
| | - J Flapper
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, CG 3584, Utrecht, the Netherlands.
| | - O Dogan
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, CG 3584, Utrecht, the Netherlands.
| | - A Khodaei
- Department of Orthopedics, University Medical Center Utrecht, Heidelberglaan 100, GA 3508, Utrecht, the Netherlands; Regenerative Medicine Center Utrecht, University Utrecht, Heidelberglaan 8, CS 3584, Utrecht, the Netherlands.
| | - G Storm
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, CG 3584, Utrecht, the Netherlands.
| | - M Croes
- Department of Orthopedics, University Medical Center Utrecht, Heidelberglaan 100, GA 3508, Utrecht, the Netherlands.
| | - M C Kruyt
- Department of Orthopedics, University Medical Center Utrecht, Heidelberglaan 100, GA 3508, Utrecht, the Netherlands; Department of Developmental Biomedical Engineering, Twente University, Drienerlolaan 5, NB 7522, Enschede, the Netherlands.
| | - D Gawlitta
- Regenerative Medicine Center Utrecht, University Utrecht, Heidelberglaan 8, CS 3584, Utrecht, the Netherlands; Department of Oral and Maxillofacial Surgery, Prosthodontics and Special Dental Care, University Medical Center Utrecht, Heidelberglaan 100, GA 3508, Utrecht, the Netherlands.
| | - H Weinans
- Department of Orthopedics, University Medical Center Utrecht, Heidelberglaan 100, GA 3508, Utrecht, the Netherlands; Department of Biomechanical Engineering, Technical University Delft, Mekelweg 2, CD 2628, Delft, the Netherlands.
| | - E Mastrobattista
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, CG 3584, Utrecht, the Netherlands.
| | - S Amin Yavari
- Department of Orthopedics, University Medical Center Utrecht, Heidelberglaan 100, GA 3508, Utrecht, the Netherlands; Regenerative Medicine Center Utrecht, University Utrecht, Heidelberglaan 8, CS 3584, Utrecht, the Netherlands; Terasaki Institute for Biomedical Innovation, 21100 Erwin St., Woodland Hills, 91367, Los Angeles, United States.
| |
Collapse
|
38
|
Huang L, Liu W, Lv X, Ge X, He Z, Yang Y, Tang Y, Wang L, Zeng J, Cheng P. Rational design, synthesis and anti-inflammatory activity of 6-substituted dihydrobenzophenanthridine derivatives. Bioorg Med Chem 2025; 122:118145. [PMID: 40056889 DOI: 10.1016/j.bmc.2025.118145] [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: 01/01/2025] [Revised: 02/21/2025] [Accepted: 02/28/2025] [Indexed: 03/10/2025]
Abstract
a series of 6-substituted dihydrobenzophenanthridine alkaloids were synthesized by introduction of different functional groups to C-6 of dihydrobenzophenanthridine backbone. The preliminary anti-inflammatory activities of all compounds were screened by investigating the inhibitory ability on NO production in LPS-stimulated RAW 264.7 cells. Among synthesized compounds, 6-(N-phenyl)-aminocarbonyl methyl dihydrochelerythrine (compound 12b) showed increased anti-inflammatory ability and decreased cytotoxicity and could inhibit the expression of pro-inflammatory factors TNF-α and IL-6 in RAW 264.7 macrophages. The anti-inflammatory ability of compound 12b was further evaluated using DSS-induced mice colitis models based on colonic tissue damage assessment, histopathological assessment and immunohistochemical analysis. In vivoexperiment revealed that compound 12b had good alleviating effect on acute colitis in mice. In conclusion, compound 12b may be a promising anti-inflammatory lead compound.
Collapse
Affiliation(s)
- Lei Huang
- Hunan Key Laboratory of Traditional Chinese Veterinary Medicine, Hunan Agricultural University, Changsha, Hunan 410128, China; Chinese Medicinal Materials Breeding Innovation Center of Yuelushan Laboratory, Changsha 410128, Hunan, China
| | - Wei Liu
- Hunan Key Laboratory of Traditional Chinese Veterinary Medicine, Hunan Agricultural University, Changsha, Hunan 410128, China; Chinese Medicinal Materials Breeding Innovation Center of Yuelushan Laboratory, Changsha 410128, Hunan, China
| | - Xinye Lv
- Hunan Key Laboratory of Traditional Chinese Veterinary Medicine, Hunan Agricultural University, Changsha, Hunan 410128, China; Chinese Medicinal Materials Breeding Innovation Center of Yuelushan Laboratory, Changsha 410128, Hunan, China
| | - Xiaomei Ge
- Hunan Key Laboratory of Traditional Chinese Veterinary Medicine, Hunan Agricultural University, Changsha, Hunan 410128, China; Chinese Medicinal Materials Breeding Innovation Center of Yuelushan Laboratory, Changsha 410128, Hunan, China
| | - Zhehao He
- Hunan Key Laboratory of Traditional Chinese Veterinary Medicine, Hunan Agricultural University, Changsha, Hunan 410128, China; Chinese Medicinal Materials Breeding Innovation Center of Yuelushan Laboratory, Changsha 410128, Hunan, China
| | - Yingxue Yang
- Hunan Key Laboratory of Traditional Chinese Veterinary Medicine, Hunan Agricultural University, Changsha, Hunan 410128, China; Chinese Medicinal Materials Breeding Innovation Center of Yuelushan Laboratory, Changsha 410128, Hunan, China
| | - Yuhui Tang
- Hunan Key Laboratory of Traditional Chinese Veterinary Medicine, Hunan Agricultural University, Changsha, Hunan 410128, China; Chinese Medicinal Materials Breeding Innovation Center of Yuelushan Laboratory, Changsha 410128, Hunan, China
| | - Lin Wang
- Hunan Key Laboratory of Traditional Chinese Veterinary Medicine, Hunan Agricultural University, Changsha, Hunan 410128, China; Chinese Medicinal Materials Breeding Innovation Center of Yuelushan Laboratory, Changsha 410128, Hunan, China
| | - Jianguo Zeng
- Hunan Key Laboratory of Traditional Chinese Veterinary Medicine, Hunan Agricultural University, Changsha, Hunan 410128, China; Chinese Medicinal Materials Breeding Innovation Center of Yuelushan Laboratory, Changsha 410128, Hunan, China.
| | - Pi Cheng
- Hunan Key Laboratory of Traditional Chinese Veterinary Medicine, Hunan Agricultural University, Changsha, Hunan 410128, China; Chinese Medicinal Materials Breeding Innovation Center of Yuelushan Laboratory, Changsha 410128, Hunan, China.
| |
Collapse
|
39
|
Oh SJ, Jang YS, Kim ER, Kim JI, Kim H, Kim Y, Park CW, Jang HS, Tran TB, Nguyen TT, Hong IK. Anti-Inflammatory Effects of Huberantha luensis and Ancistrocladus tectorius Extracts Through Inhibition of Nuclear Factor-κB and Mitogen-Activated Protein Kinase Pathways. J Med Food 2025; 28:448-454. [PMID: 40256789 DOI: 10.1089/jmf.2024.k.0276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/22/2025] Open
Abstract
Inflammation is a crucial response to harmful stimuli, but its chronic activation contributes to various diseases, including inflammatory bowel disease, osteoarthritis, and neurological disorders. While nonsteroidal anti-inflammatory drugs are widely used as anti-inflammation drugs, their extended usage often results in severe side effects, emphasizing the need for safer alternatives. Therefore, it is of the greatest importance to identify and discover new anti-inflammatory agents that exhibit a reduced incidence of adverse side effects. This study investigates the anti-inflammatory potential of methanol extracts from eight native Vietnamese plant species. These extracts were screened for their ability to inhibit nitric oxide production and pro-inflammatory cytokine expression in lipopolysaccharides-stimulated RAW264.7 macrophages. Among the tested extracts, those derived from Huberantha luensis (Pierre) Chaowasku and Ancistrocladus tectorius (Lour.) Merr. demonstrated notable reductions in NO, TNF-α, interleukin (IL)-1β, and IL-6 levels. Further analysis revealed that these extracts are abundant in polyphenols and flavonoids, compounds recognized for their anti-inflammatory effects. Furthermore, these extracts exerted their effects by inhibiting the kappa-light-chain-enhancer of activated B cells and mitogen-activated protein kinase signaling pathways, as evidenced by reduced phosphorylation of the proteins. These results suggest that the methanol extracts obtained from H. luensis and A. tectorius possess considerable potential in paving the way towards the innovative development of new therapeutic approaches aimed at alleviating chronic inflammation.
Collapse
Affiliation(s)
| | - Young Sun Jang
- Kangwon National University College of Pharmacy, Chuncheon, Korea
| | | | | | | | | | | | | | - The Bach Tran
- Vietnam Academy of Science and Technology Institute of Ecology and Biological Resources, Hanoi, Vietnam
| | - Thu Thuy Nguyen
- Korea Research Institute of Bioscience & Biotechnology, Biological Material Research Center, Yuseong-gu, Korea
| | | |
Collapse
|
40
|
Nie P, Hu L, You T, Jia T, Xu H. Lead Mediated Lipopolysaccharides Exacerbates Fatty Liver Processes in High-Fat Diets-Induced Mice. ENVIRONMENTAL TOXICOLOGY 2025; 40:750-763. [PMID: 39715151 DOI: 10.1002/tox.24463] [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: 03/05/2024] [Revised: 10/10/2024] [Accepted: 12/08/2024] [Indexed: 12/25/2024]
Abstract
Obesity leads to a variety of health risks, and lead, which is ranked second in Agency for Toxic Substances and Disease Registry's priority list of harmful substances, may be more harmful to individuals that are obese. C57BL/6 mice were fed a normal diet or a high-fat diet with or without exposure to 1 g/L lead exposure in drinking water for 8 consecutive weeks. Serum and hepatic biochemistry analysis, histopathological observation, and RT-qPCR were used to explore the potential mechanism of liver damage in obese individuals after Pb exposure, and fecal microbiota transplantation was performed to investigate the role of the gut microbiota in the progression of fatty liver disease. We found that the progression of fatty liver disease induced by high-fat diets was accelerated by chronic lead intake. In addition, the occurrences of liver injury in recipient mice suggested the role of the gut microbiota. These findings indicated that the combination of lead and a HFD exacerbated hepatic lipotoxicity by activating LPS-mediated inflammation, and that gut microbiota disorders and impaired intestinal barrier function play pivotal roles in the progression of fatty liver disease.
Collapse
Affiliation(s)
- Penghui Nie
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, China
| | - Liehai Hu
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, China
| | - Tao You
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, China
| | - Tiantian Jia
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, China
| | - Hengyi Xu
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, China
- International Institute of Food Innovation co. Ltd., Nanchang University, Nanchang, People's Republic of China
| |
Collapse
|
41
|
Agrawal N. A Comprehensive Review on the Advancements of Dual COX-2/5-LOX Inhibitors as Anti-Inflammatory Drugs. Chem Biol Drug Des 2025; 105:e70114. [PMID: 40346930 DOI: 10.1111/cbdd.70114] [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: 02/19/2025] [Revised: 04/03/2025] [Accepted: 04/15/2025] [Indexed: 05/12/2025]
Abstract
Chronic pain and inflammation are widespread clinical issues that significantly affect patients' quality of life and are often associated with serious conditions such as arthritis, cancer, and cardiovascular disease. Effective management of inflammation is therefore a major public health priority. Current anti-inflammatory treatments-including non-steroidal anti-inflammatory drugs (NSAIDs), corticosteroids, disease-modifying antirheumatic drugs (DMARDs), and biologics-offer symptomatic relief but are frequently limited by side effects such as gastrointestinal toxicity, immunosuppression, or cardiovascular risks. Moreover, most of these therapies target only a single pathway in the inflammatory cascade. Dual inhibitors of COX-2 and 5-LOX have emerged as a promising therapeutic class, as they simultaneously block two key enzymes involved in prostaglandin and leukotriene synthesis. This dual-action approach offers enhanced efficacy and may reduce adverse effects linked to selective or non-selective COX inhibition. This review discusses the underlying mechanisms of inflammation, evaluates current treatment options, and highlights the pharmacological advantages and development status of dual COX-2/5-LOX inhibitors as a next-generation strategy for inflammation management.
Collapse
Affiliation(s)
- Neetu Agrawal
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India
| |
Collapse
|
42
|
Mingels S. Paediatric pain and malnutrition in low-income countries: A narrative review. Nutr Health 2025:2601060251336823. [PMID: 40304640 DOI: 10.1177/02601060251336823] [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: 05/02/2025]
Abstract
Background: Despite its omnipresence, paediatric pain remains poorly understood and documented, especially in low-income countries. Such pain can be a symptom of long-term subclinical conditions such as systemic chronic inflammation (SCI). The latter can be related to malnutrition. Aim: To explore a potential association between paediatric pain and malnutrition in low-income countries. Methods: Narrative review, including a literature search in the PubMed, EMBASE, Web of Science and Scopus databases (update 24 March 2025). The search query comprised controlled terminology and free text words relating to 'Malnutrition', 'Pain', 'SCI' and 'Paediatric'. Results: To comprehend the complex relation between malnutrition and paediatric pain, associations between (1a) malnutrition, and nociceptive brain development, (1b) malnutrition, the gut microbiome and SCI, and (2) SCI and pain were explored. (1a) Early noxious exposure (e.g. malnutrition-related SCI) can cause long-term alterations in pain perception, brain function and structures. The consequences of malnutrition on the nociceptive brain depend on the life-cycle. (1b) Moderate acute malnutrition causes chronic inflammation and exaggerated inflammatory responses. Such responses could indicate hyper-inflammatory phenotypes. (2) Systemic-induced inflammation causes a widespread increase in musculoskeletal pain sensitivity. Conclusion: Malnutrition could contribute to the development of a nociceptive brain and SCI. Malnutrition-related SCI could induce changes in pain perception/thresholds, and predispose to developing chronic pain. If a relation between malnutrition and SCI predisposes children to develop pain, the prevailing biophysical approach needs revision. A multidimensional interdisciplinary approach seems more relevant. Such approach includes social, cognitive, socioeconomic, lifestyle, nutritional (e.g. integrating nutritional and microbiome-targeted interventions) and environmental dimensions.
Collapse
Affiliation(s)
- Sarah Mingels
- Musculoskeletal Research Unit, Department of Rehabilitation Sciences, Faculty of Movement and Rehabilitation Sciences, Leuven University, Leuven, Belgium
| |
Collapse
|
43
|
Ahmadi N, Rincón M, Mallandrich M, Suñer-Carbó J, Sosa L, Zelaya M, Martinez-Ruiz S, Cordero C, Calpena AC. Novel Gels for Post-Piercing Care: Evaluating the Efficacy of Pranoprofen Formulations in Reducing Inflammation. Gels 2025; 11:334. [PMID: 40422354 DOI: 10.3390/gels11050334] [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: 02/18/2025] [Revised: 04/17/2025] [Accepted: 04/21/2025] [Indexed: 05/28/2025] Open
Abstract
Mild to moderate pain for a few hours to several days post-piercing is normal, and the pain is usually accompanied by swelling, redness, and warmth due to the inflammatory response. Cool compresses and over-the-counter analgesics (e.g., NSAIDs) can ease mild discomfort. However, oral NSAIDs may have systemic side effects; for this reason, we propose a topical anti-inflammatory approach. Four pranoprofen-loaded gels were created using different gelling agents: Sepigel® 305 (PF-Gel-Sep), Carbopol® 940 (PF-Gel-Car), Pluronic® F-68 (PF-Gel-Plu), and Lutrol® F-127 (PF-Gel-Lut). The gels were assessed for pH, morphology, FT-IR spectroscopy, rheological properties, spreadability, swelling and degradation, drug release kinetics, skin permeation (cow and human skin), irritation potential (HET-CAM assay), and impact on skin barrier function (TEWL and SCH). The gels exhibited varied rheological properties with PF-Gel-Car showing high viscosity and PF-Gel-Plu very low viscosity. All gels had similar spreadability with PF-Gel-Lut showing the highest. PF-Gel-Car showed the highest amounts of PF released, whereas PF-Gel-Plu led to the highest amount of pranoprofen retained in human and bovine skin. The HET-CAM assay indicated that none of the PF-Gels were irritating. Additionally, PF-Gel-Car and PF-Gel-Plu showed no cytotoxic effects on HaCaT cells. In vivo testing on mice showed that PF-Gel-Car prevented inflammation, while the rest of the gels were able to revert it in 25 min. Skin tolerance tests revealed the gels did not affect TEWL, and some gels improved SCH. The study successfully formulated and characterized four PF-loaded topical gels with potential to be used as an alternative for treating inflammation from piercings and ear tags.
Collapse
Affiliation(s)
- Negar Ahmadi
- Department of Pharmacy, Pharmaceutical Technology and Physical Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Av. Joan XXIII 27-31, 08028 Barcelona, Spain
| | - Maria Rincón
- Department of Materials Science and Physical Chemistry, Faculty of Chemistry, University of Barcelona, C. Martí i Franquès 1-11, 08028 Barcelona, Spain
| | - Mireia Mallandrich
- Department of Pharmacy, Pharmaceutical Technology and Physical Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Av. Joan XXIII 27-31, 08028 Barcelona, Spain
- Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, Diagonal Ave. 645, 08028 Barcelona, Spain
| | - Joaquim Suñer-Carbó
- Department of Pharmacy, Pharmaceutical Technology and Physical Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Av. Joan XXIII 27-31, 08028 Barcelona, Spain
- Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, Diagonal Ave. 645, 08028 Barcelona, Spain
| | - Lilian Sosa
- Centro Experimental en Biociencia (CENBIO), Facultad de Ciencias Químicas y Farmacia, Universidad Nacional Autónoma de Honduras (UNAH), Tegucigalpa 11101, Honduras
- Instituto de Investigaciones en Microbiología (IIM), Facultad de Ciencias, Universidad Nacional Autónoma de Honduras (UNAH), Tegucigalpa 11101, Honduras
| | - Mireya Zelaya
- Laboratorio de Técnicas Histológicas, Facultad de Ciencias, Universidad Nacional Autónoma de Honduras (UNAH), Tegucigalpa 11101, Honduras
| | - Sergio Martinez-Ruiz
- Departament de Bioquímica i Fisiologia, Facultat de Farmàcia i Ciències de l'Alimentació, University of Barcelona, 08028 Barcelona, Spain
| | - Cecilia Cordero
- Departament de Bioquímica i Fisiologia, Facultat de Farmàcia i Ciències de l'Alimentació, University of Barcelona, 08028 Barcelona, Spain
| | - Ana C Calpena
- Department of Pharmacy, Pharmaceutical Technology and Physical Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Av. Joan XXIII 27-31, 08028 Barcelona, Spain
- Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, Diagonal Ave. 645, 08028 Barcelona, Spain
| |
Collapse
|
44
|
Zhang Y, Yue Y, Sun Z, Li P, Wang X, Cheng G, Huang H, Li Z. Pan-immune-inflammation value and its association with all-cause and cause-specific mortality in the general population: a nationwide cohort study. Front Endocrinol (Lausanne) 2025; 16:1534018. [PMID: 40370772 PMCID: PMC12074934 DOI: 10.3389/fendo.2025.1534018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 04/10/2025] [Indexed: 05/16/2025] Open
Abstract
Introduction The Pan-Immune-Inflammation Value (PIV) is a novel biomarker derived from counts of neutrophils, platelets, monocytes, and lymphocytes, providing a comprehensive measure of systemic immune and inflammatory status. While it has shown prognostic value in specific disease settings, its association with mortality in the general population remains unclear. This study aims to evaluate the predictive value of PIV for all-cause and cause-specific mortality, including cardiovascular, cancer, and diabetes-related deaths, within a general adult population. Methods Data were obtained from the NHANES cohort, with 48,662 participants aged 20 and older. Participants were followed for an average of 117.44 months, with PIV quartiles calculated at baseline. Cox proportional hazard models were used to assess mortality risk across PIV quartiles, while restricted cubic spline models examined nonlinear dose-response relationships. Subgroup and sensitivity analyses further explored the robustness of PIV's associations. Results Higher PIV levels were significantly associated with increased risks of all-cause, cardiovascular, cancer, and diabetes mortality. Nonlinear relationships were observed between PIV and all-cause, cardiovascular, and cancer mortality, with a risk threshold at PIV values above 254.07. Subgroup analyses supported these findings, and sensitivity analyses confirmed the consistency of PIV's prognostic value. Conclusion Elevated PIV serves as an independent risk factor for multiple mortality outcomes in the general population. This study underscores the potential of PIV as a predictive biomarker for mortality risk, with implications for its use in clinical and epidemiological settings. Further studies are needed to confirm PIV's clinical utility across diverse populations and conditions.
Collapse
Affiliation(s)
- Ye Zhang
- Department of Neurosurgery, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, Sichuan, China
| | - Yong Yue
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, Japan
- Department of Pharmacology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Zhengyu Sun
- Department of Plastic and Aesthetic, Jintang First People’s Hospital, Chengdu, Sichuan, China
| | - Pengcheng Li
- The Department of Oncology, The First Affiliated Hospital of the Chengdu Medical College, Chengdu, Sichuan, China
| | - Xiaoyi Wang
- Department of Neurosurgery, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, Sichuan, China
| | - Gang Cheng
- Department of Neurosurgery, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, Sichuan, China
| | - Hailin Huang
- Department of Neurosurgery, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, Sichuan, China
| | - Zongping Li
- Department of Neurosurgery, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, Sichuan, China
| |
Collapse
|
45
|
Varol C, Fındık Y, Baykul T. Evaluation of the effect of the neutrophil-to-lymphocyte ratio on facial oedema after orthognathic surgery: a 12-month follow-up study. Int J Oral Maxillofac Surg 2025:S0901-5027(25)00117-1. [PMID: 40307074 DOI: 10.1016/j.ijom.2025.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 02/04/2025] [Accepted: 04/08/2025] [Indexed: 05/02/2025]
Abstract
Orthognathic surgery to correct dentofacial deformities has been performed safely for many years. Postoperative facial oedema occurs to varying degrees in almost all patients. The aim of this study was to evaluate the relationship between the preoperative neutrophil-to-lymphocyte ratio (NLR) and the severity of postoperative facial oedema using 3D stereophotogrammetry. Thirty patients (Le Fort I + BSSO) were included in this study. They were divided into two groups according to their NLR: ≥2.00 (n = 13) and <2.00 (n = 17). Patients were followed for 12 months and facial oedema was measured on days 1, 2, 3 and 10, and months 1 and 6, using the 12-month image as the reference. Postoperative facial oedema was significantly greater in the NLR ≥2.00 group in the first 10 days (days 1, 2, 3, 10: P = 0.011, P = 0.010, P = 0.040, P = 0.032), while no significant difference was observed for the long-term measurements (months 1, 6: P = 0.693, P = 0.374). The results of this study suggest that NLR may serve as a potential biomarker to predict the severity of postoperative facial oedema and may assist clinicians in their decision-making process when managing the postoperative inflammatory response.
Collapse
Affiliation(s)
- C Varol
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Suleyman Demirel University, Isparta, Turkey.
| | - Y Fındık
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Suleyman Demirel University, Isparta, Turkey.
| | - T Baykul
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Suleyman Demirel University, Isparta, Turkey.
| |
Collapse
|
46
|
Gao J, Li W, Lin J, Han Y, Ji G, Liu Z. Galnt3, an enzyme engaged in protein glycosylation modification, is essential for the maintaining of intestinal health in zebrafish. FISH & SHELLFISH IMMUNOLOGY 2025; 163:110373. [PMID: 40306380 DOI: 10.1016/j.fsi.2025.110373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 04/26/2025] [Accepted: 04/27/2025] [Indexed: 05/02/2025]
Abstract
Intestinal inflammation significantly impairs intestinal function and is closely associated with various health complications. Understanding its molecular mechanisms is crucial for developing effective therapeutic strategies. Galnt3, a member of the polypeptide N-acetylgalactosaminyltransferase family, participates in multiple biological processes, yet its specific role in intestinal inflammation remains poorly understood. In this study, we observed a significant downregulation of zebrafish galnt3 in response to GCRV virus or poly(I:C) infection. Galnt3 knockout (galnt3-/-) zebrafish exhibited reduced survival rates, particularly following GCRV virus inoculation, accompanied by severe ascites and abdominal hemorrhage. Histopathological examination of intestinal tissues revealed thinning of intestinal walls, shortened villi, and increased acidic mucus secretion, all indicative of aggravated intestinal inflammation. Furthermore, galnt3 deficiency was found to trigger the upregulation of numerous pro-inflammatory cytokine genes. Through cell scratch assays and p38 MAPK phosphorylation analysis, we demonstrated that Galnt3 inhibits p38 MAPK phosphorylation and macrophage migration, thereby reducing the production of pro-inflammatory factors. Our findings highlight the pivotal role of Galnt3 in maintaining intestinal homeostasis and regulating inflammatory responses, providing valuable insights into the molecular mechanisms underlying intestinal inflammation and identifying potential therapeutic targets.
Collapse
Affiliation(s)
- Jing Gao
- College of Marine Life Sciences, Key Laboratory of Evolution & Marine Biodiversity (Ministry of Education) and Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao, 266003, China
| | - Wenjin Li
- College of Marine Life Sciences, Key Laboratory of Evolution & Marine Biodiversity (Ministry of Education) and Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao, 266003, China
| | - Jingyuan Lin
- College of Marine Life Sciences, Key Laboratory of Evolution & Marine Biodiversity (Ministry of Education) and Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao, 266003, China
| | - Yilin Han
- College of Marine Life Sciences, Key Laboratory of Evolution & Marine Biodiversity (Ministry of Education) and Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao, 266003, China
| | - Guangdong Ji
- College of Marine Life Sciences, Key Laboratory of Evolution & Marine Biodiversity (Ministry of Education) and Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao, 266003, China; Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, 266237, China
| | - Zhenhui Liu
- College of Marine Life Sciences, Key Laboratory of Evolution & Marine Biodiversity (Ministry of Education) and Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao, 266003, China; Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, 266237, China.
| |
Collapse
|
47
|
González-Hernández M, Gallardo-Andalucía L, Hernansanz-Agustín P. Modes of Mitochondrial Reactive Oxygen Species Production in Inflammation. Antioxid Redox Signal 2025. [PMID: 40285481 DOI: 10.1089/ars.2024.0737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/29/2025]
Abstract
Background: Inflammation is one of the most important pathways in innate immunity and its relationship with redox biology is becoming increasingly clear in the last decades. However, the specific redox modes and pathways by which inflammation is produced are not yet well defined. Significance: In this review, we provide a general explanation of the reactive oxygen species (ROS) production and quenching modes occurring in mammalian mitochondria, as well as a summary of the most recent advances in mitochondrial redox biology and bioenergetics regarding sodium (Na+) homeostasis. In addition, we provide a collection of examples in which several inflammatory pathways have been associated with specific modes of either mitochondrial ROS production or quenching. Innovation: The role of Na+ in mitochondrial biology is being developed. Since its discovery as a second messenger, the research of its role in the immune system has emerged. Now, the role of Na+ in mitochondrial bioenergetics has recently been identified, which owns unprecedented applications. The potential implication of Na+ in inflammatory mechanisms grows as its role does not only cover ROS production and respiration but also the control through the management of mitochondrial membrane potential. Future directions: Na+ is becoming relevant for mitochondrial biology. Thus, processes regarding mitochondrial bioenergetics, redox state, or metabolism may probably need to include the study of Na+ in their road map. Some of these pathways are involved in inflammation and more are possibly to come. This review is expected to serve as a bridge between both fields. Antioxid. Redox Signal. 00, 000-000.
Collapse
Affiliation(s)
- Miguel González-Hernández
- Departamento de Neurobiología Molecular, Celular y del Desarrollo, Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | | | - Pablo Hernansanz-Agustín
- Departamento de Neurobiología Molecular, Celular y del Desarrollo, Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| |
Collapse
|
48
|
Rosell A, Krygowska AA, Alcón Pérez M, Cuesta C, Voisin MB, de Paz J, Sanz-Fraile H, Rajeeve V, Carreras-González A, Berral-González A, Swinyard O, Gabandé-Rodríguez E, Downward J, Alcaraz J, Anguita J, García-Macías C, De Las Rivas J, Cutillas PR, Castellano Sanchez E. RAS-p110α signalling in macrophages is required for effective inflammatory response and resolution of inflammation. eLife 2025; 13:RP94590. [PMID: 40272400 PMCID: PMC12021417 DOI: 10.7554/elife.94590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2025] Open
Abstract
Macrophages are crucial in the body's inflammatory response, with tightly regulated functions for optimal immune system performance. Our study reveals that the RAS-p110α signalling pathway, known for its involvement in various biological processes and tumourigenesis, regulates two vital aspects of the inflammatory response in macrophages: the initial monocyte movement and later-stage lysosomal function. Disrupting this pathway, either in a mouse model or through drug intervention, hampers the inflammatory response, leading to delayed resolution and the development of more severe acute inflammatory reactions in live models. This discovery uncovers a previously unknown role of the p110α isoform in immune regulation within macrophages, offering insight into the complex mechanisms governing their function during inflammation and opening new avenues for modulating inflammatory responses.
Collapse
Affiliation(s)
- Alejandro Rosell
- Tumour-Stroma Signalling Lab., Centro de Investigación del Cáncer, Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad de Salamanca, Campus Miguel de UnamunoSalamancaSpain
| | - Agata Adelajda Krygowska
- Centre for Cancer and Inflammation, Barts Cancer Institute, Queen Mary University of LondonLondonUnited Kingdom
| | - Marta Alcón Pérez
- Tumour-Stroma Signalling Lab., Centro de Investigación del Cáncer, Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad de Salamanca, Campus Miguel de UnamunoSalamancaSpain
| | - Cristina Cuesta
- Tumour-Stroma Signalling Lab., Centro de Investigación del Cáncer, Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad de Salamanca, Campus Miguel de UnamunoSalamancaSpain
| | - Mathieu-Benoit Voisin
- Centre for Microvascular Research, William Harvey Research Institute, Queen Mary University of LondonLondonUnited Kingdom
| | - Juan de Paz
- Tumour-Stroma Signalling Lab., Centro de Investigación del Cáncer, Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad de Salamanca, Campus Miguel de UnamunoSalamancaSpain
| | - Héctor Sanz-Fraile
- Unit of Biophysics and Bioengineering, Department of Biomedicine, School of Medicine and Health Sciences, Universitat de BarcelonaBarcelonaSpain
| | - Vinothini Rajeeve
- Centre for Cancer Genomics and Computational Biology, Cell Signalling and Proteomics Laboratory, Barts Cancer Institute, Queen Mary University of LondonLondonUnited Kingdom
| | - Ana Carreras-González
- Bioinformatics and Functional Genomics, Centro de Investigación del Cáncer, Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad de SalamancaSalamancaSpain
| | | | - Ottilie Swinyard
- Centre for Cancer and Inflammation, Barts Cancer Institute, Queen Mary University of LondonLondonUnited Kingdom
| | - Enrique Gabandé-Rodríguez
- Centre for Cancer and Inflammation, Barts Cancer Institute, Queen Mary University of LondonLondonUnited Kingdom
| | - Julian Downward
- Oncogene Biology Laboratory, Francis Crick InstituteLondonUnited Kingdom
| | - Jordi Alcaraz
- Unit of Biophysics and Bioengineering, Department of Biomedicine, School of Medicine and Health Sciences, Universitat de BarcelonaBarcelonaSpain
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology (BIST)BarcelonaSpain
| | - Juan Anguita
- Inflammation and Macrophage Plasticity Lab, CIC bioGUNEDerioSpain
- Ikerbasque, Basque Foundation for ScienceBilbaoSpain
- Pathology Unit, Centro de Investigación del Cáncer, Instituto de Biología Molecular y Celular del Cáncer, Universidad de SalamancaSalamancaSpain
| | - Carmen García-Macías
- Pathology Unit, Centro de Investigación del Cáncer, Instituto de Biología Molecular y Celular del Cáncer, Universidad de SalamancaSalamancaSpain
| | - Javier De Las Rivas
- Bioinformatics and Functional Genomics, Centro de Investigación del Cáncer, Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad de SalamancaSalamancaSpain
| | - Pedro R Cutillas
- Centre for Cancer Genomics and Computational Biology, Cell Signalling and Proteomics Laboratory, Barts Cancer Institute, Queen Mary University of LondonLondonUnited Kingdom
| | - Esther Castellano Sanchez
- Tumour-Stroma Signalling Lab., Centro de Investigación del Cáncer, Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad de Salamanca, Campus Miguel de UnamunoSalamancaSpain
- Centre for Cancer and Inflammation, Barts Cancer Institute, Queen Mary University of LondonLondonUnited Kingdom
| |
Collapse
|
49
|
Tang Y, Guo T, Wang X, Li C, Zhang X, Zhang J. Cyclodextrin-Derived Macromolecular Therapies for Inflammatory Diseases. Macromol Biosci 2025:e2400637. [PMID: 40271896 DOI: 10.1002/mabi.202400637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 04/02/2025] [Indexed: 04/25/2025]
Abstract
Inflammation is an essential physiological defense mechanism against harmful stimuli, yet dysregulated inflammatory responses are closely associated with the pathogenesis of numerous acute and chronic diseases. Recent advances highlight the remarkable anti-inflammatory potential of bioactive macromolecules, particularly cyclodextrins (CDs) and their engineered derivatives, which are emerging as promising therapeutic agents. This review systematically introduces different CDs and CD-derived macromolecules that demonstrate anti-inflammatory properties, with emphasis on their molecular mechanisms of action. Native CDs exhibit direct therapeutic effects through host-guest interactions, enabling selective sequestration of pathogenic components such as cholesterol crystals and proteins that drive inflammatory cascades. Moreover, chemically modified CD derivatives incorporating functional groups demonstrate enhanced capabilities in neutralizing inflammatory mediators and modulating immune cell responses. This work further discusses the expanding therapeutic applications of these macromolecules across diverse inflammatory conditions, ranging from acute tissue injuries to chronic autoimmune disorders. Finally, this work critically analyzes the crucial challenges and emerging opportunities in translating CD-based macromolecular therapies into clinical practice, addressing key considerations in biocompatibility, targeted delivery, and therapeutic efficacy optimization.
Collapse
Affiliation(s)
- Yige Tang
- College of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
- International Medical College, Chongqing Medical University, Chongqing, 400016, China
| | - Tao Guo
- College of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
| | - Xuanran Wang
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Chenwen Li
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Xiangjun Zhang
- College of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
| | - Jianxiang Zhang
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Yu-Yue Pathology Scientific Research Center, 313 Gaoteng Avenue, Jiulongpo District, Chongqing, 400039, China
| |
Collapse
|
50
|
Zhou ZY, Zhou ZP, Yue YX, Zhong YK, Yang ZX, Cai GL. Dimethyl Pent-2-Enedioate inhibits LPS-induced inflammatory response in macrophages. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2025:S1684-1182(25)00084-2. [PMID: 40300966 DOI: 10.1016/j.jmii.2025.03.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 03/01/2025] [Accepted: 03/27/2025] [Indexed: 05/01/2025]
Abstract
BACKGROUND Endogenous metabolite itaconate and its derivative Dimethyl itaconate (DMI) exhibit significant anti-inflammatory effects. Dimethyl Pent-2-Enedioate (DMP), an isomer of DMI, may possess similar properties. This study investigates the anti-inflammatory effects of DMP in LPS-induced macrophages and explores its potential regulatory mechanisms. METHODS Inflammatory marker levels were assessed at both the mRNA and protein levels using ELISA and qRT-PCR. The activation status of macrophages was evaluated by flow cytometry, quantifying the number of CD40-positive cells. RNA sequencing was conducted to investigate the transcriptomic changes following DMP treatment. Subsequent GO and KEGG enrichment analyses were performed to identify potential mechanisms underlying DMP's effects. Western blot analysis was employed to assess the expression of p-p65, while immunofluorescence analysis was used to examine p65 nuclear translocation, providing insight into the regulatory effects of DMP on the NF-κB signaling pathway. RESULTS DMP inhibited the expression of inflammatory markers TNF-α, IL-6, and MCP-1 at both mRNA and protein levels. Flow cytometry analysis revealed a decrease in CD40-positive cells. RNA sequencing identified DEGs enriched in inflammation-related pathways. Western blotting and immunofluorescence confirmed that DMP reduced p-p65 expression and inhibited p65 nuclear translocation, suggesting a potential regulatory effect on the NF-κB signaling pathway. CONCLUSION DMP significantly inhibits LPS-induced inflammation in macrophages, with its underlying mechanisms being complex. Our data demonstrate that DMP exerts its anti-inflammatory effects at least in part through the downregulation of the NF-κB pathway, offering potential applications in the prevention and treatment of inflammation-related diseases.
Collapse
Affiliation(s)
- Zhi-Ying Zhou
- Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China; Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Zhi-Peng Zhou
- University of Chinese Academy of Sciences, Beijing, China
| | - Ying-Xing Yue
- Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Yu-Ke Zhong
- Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Zhou-Xin Yang
- Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China.
| | - Guo-Long Cai
- Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China.
| |
Collapse
|