1
|
Li J, Li L, Liang W, Li L, Wang R, Wang Z, Ma C. Spatial multi-omics analysis of metabolic heterogeneity in zebrafish exposed to microcystin-LR and its disinfection byproducts. WATER RESEARCH 2025; 280:123599. [PMID: 40209558 DOI: 10.1016/j.watres.2025.123599] [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: 01/15/2025] [Revised: 03/16/2025] [Accepted: 04/04/2025] [Indexed: 04/12/2025]
Abstract
Most studies on the biological effects of exogenous pollutants have focused on whole samples or cell populations, and lack spatial heterogeneity consideration due to technical limitations. Microcystin-LR (MC-LR) from cyanobacterial blooms threatens ecosystems and human health, while microcystin-LR disinfection by-products (MCLR-DBPs) in drinking water remain a concern for their toxin-like structure. This study introduces spatial multi-omics to investigate the disruptions caused by ingestion of MC-LR and MCLR-DBPs in zebrafish. The method integrates metabolomics, spatial metabolomics, and spatial transcriptomics to characterize the overall metabolic changes in whole zebrafish caused by MC-LR and MCLR-DBPs, then provides further insight into the variation of spatial distribution of metabolites and genes in MC-LR and MCLR-DBPs targeted organ. The results showed that MC-LR and MCLR-DBPs induced oxidative stress and metabolic imbalance, and disrupted the physiological homeostasis of zebrafish. Spatial multi-omics analysis further revealed that MC-LR and MCLR-DBPs exacerbate disruptions in energy and lipid metabolism, methylation processes, and immune pathways by modulating the expression of genes such as gatm, gnmt, cyp2p9, and tdo2b. In conclusion, this study developed a spatial multi-omics approach that not only enhances the understanding of the biological effects of MC-LR and MCLR-DBPs but also provides robust technical support for investigating other environmental pollutants.
Collapse
Affiliation(s)
- Jun Li
- Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, PR China
| | - Lili Li
- Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, PR China
| | - Weiqiang Liang
- The First Affiliated Hospital of Shandong First Medical University, Shandong First Medical University, Jinan, Shandong, 250014, PR China
| | - Lingyu Li
- Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, PR China; College of Food Science and Engineering, Shandong Agricultural University, Tai'an, 271018, PR China
| | - Ruya Wang
- Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, PR China; School of Pharmaceutical Sciences, Jilin University, Changchun 130021, PR China
| | - Zhenhua Wang
- Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, PR China.
| | - Chunxia Ma
- Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, PR China; State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 1007002, PR China.
| |
Collapse
|
2
|
Chen L, Feng J, Wang G, Liu S. Therapeutic potential of Platycodin D in allergic asthma through anti-inflammatory and anti-remodeling effects. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 142:156728. [PMID: 40397997 DOI: 10.1016/j.phymed.2025.156728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 03/31/2025] [Accepted: 04/02/2025] [Indexed: 05/23/2025]
Abstract
BACKGROUND Allergic asthma (AA) is a prevalent chronic respiratory disease characterized by airway hyperresponsiveness (AHR) and chronic inflammation, significantly impairing patients' quality of life. PURPOSE This study investigates the therapeutic effects of Platycodin D (PLD) on AA and its underlying mechanisms via the EGFR/PI3K/Akt signaling pathway. METHODS In vitro, BEAS-2B cells treated with IL-4 and IL-13 simulated asthma's inflammatory environment. Enzyme-linked immunosorbent assay (ELISA) assessed PLD's modulation of inflammatory factors, while Western blot (WB) analyzed its impact on airway remodeling proteins. Induced pluripotent stem cells (iPSC)-derived airway organoids (AOs) were used to evaluate PLD's effects on airway remodeling, observed through tissue staining and immunofluorescence. In vivo, an OVA-induced asthma mouse model was employed to assess PLD's therapeutic potential via lung function tests, serum biochemical analysis, and histopathology. Network pharmacology and transcriptomics predicted and validated PLD's target pathways. RESULTS In vivo experiments demonstrated that PLD significantly alleviated airway inflammation and remodeling in OVA-induced asthmatic mice. Specifically, treatment with 5 mg/kg PLD significantly reduced the number of inflammatory cells recovered from bronchoalveolar lavage fluid (BALF) compared to the model group (p < 0.05). Serum levels of IgE, IL-4, IL-5, IL-13, and IL-17A were markedly decreased following PLD treatment (p < 0.05). PLD also improved lung function by reducing airway resistance (RL) across all tested methacholine concentrations, with significant reductions at 5, 10, and 20 mg/mL doses (p < 0.05). Histological analysis revealed that PLD attenuated pathological changes in lung tissues, including goblet cell hyperplasia and collagen deposition. Western blot analysis confirmed that PLD significantly downregulated the expression of COL1A1 and α-SMA in lung tissues (p < 0.05), suggesting suppression of airway remodeling. In vitro, PLD inhibited the expression of IL-6, IL-8, COL1A1, and α-SMA in human bronchial epithelial (HBE) cells in a dose-dependent manner. Transcriptomic sequencing and RT-qPCR analysis further demonstrated that PLD downregulated key genes involved in the EGFR/PI3K/Akt pathway. Molecular docking showed high binding affinity between PLD and EGFR/PI3K proteins, supporting a potential mechanistic link. CONCLUSION PLD exerts therapeutic effects in allergic asthma by suppressing airway inflammation, improving lung function, and inhibiting airway remodeling. These effects are associated with the inhibition of the EGFR/PI3K/Akt signaling pathway. Our findings suggest that PLD may serve as a promising candidate for the treatment of allergic airway diseases.
Collapse
Affiliation(s)
- Li Chen
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Jianwei Feng
- Department of Ultrasound, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Guina Wang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| | - Si Liu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| |
Collapse
|
3
|
Gu J, Ruan J, Guo C, Li Z, Fu H, Xie Y, Xie H, Gong X, Shi H. Organelles Ca 2+ redistribution contributes to cadmium-induced EMT of renal cancer cells through p-cPLA 2-mediated arachidonic acid release. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 298:118317. [PMID: 40383071 DOI: 10.1016/j.ecoenv.2025.118317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 04/28/2025] [Accepted: 05/10/2025] [Indexed: 05/20/2025]
Abstract
Cadmium ion (Cd2+) is a non-essential metal that can increase cancer risk, including potentially renal cell carcinoma (RCC), though this link is not definitive. Cd2+ exposure impairs fatty acid metabolism in the kidneys, particularly affecting arachidonic acid (AA) levels, which are crucial for health. Previous studies have suggested that Cd2+-altered the AA metabolism associates with renal dysfunction. However, the role and mechanism of Cd2+-regulated AA source in promoting RCC progression are still unclear. This study aims to investigate how Cd2+ exposure affects AA levels in renal cancer cells and its role in promoting cell migration. Cd2+ exposure increases AA levels through cPLA2-mediated release. It also induces calcium ion (Ca2+) redistribution from the endoplasmic reticulum (ER) to mitochondria, activating the p38 MAPK/cPLA2 signaling pathway, and epithelial-mesenchymal transition (EMT) of Caki-1 cells. Cd2+-induced ER Ca2+ release, p38 MAPK/cPLA2 signaling activation, AA levels, and EMT of Caki-1 cells were effectively reversed by siRNA knockdown of IP3R. Both exogenous AA treatments and Cd2+-induced AA metabolite PGD2 promoted EMT and cell migration of Caki-1 cells. This study highlights Cd2+'s impact on fatty acid metabolism and organelle function in renal cancer cells, identifying potential therapeutic targets for RCC.
Collapse
Affiliation(s)
- Jie Gu
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu 212000, China.
| | - Jiacheng Ruan
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu 212000, China
| | - Chuanzhi Guo
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu 212000, China
| | - Zehua Li
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu 212000, China
| | - Huilin Fu
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu 212000, China
| | - Yimin Xie
- Affiliated Hospital of Jiangsu University-Yixing Hospital, Yixing, Jiangsu 214200, China
| | - Hebing Xie
- Jiangsu Shenhou Pharmaceutical Research Co., Ltd., Nantong, 226133, China
| | - Xun Gong
- Department of Rheumatology & Immunology, Affiliated Hospital of Jiangsu University, Zhenjiang 212013, China
| | - Haifeng Shi
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu 212000, China.
| |
Collapse
|
4
|
Lengerli D, Bakht A, Çalışkan K, Dahlke P, Bal NB, Jordan PM, Çalışkan B, Werz O, Banoglu E. Phenyl-benzyl-ureas with pyridazinone motif: Potent soluble epoxide hydrolase inhibitors with enhanced pharmacokinetics and efficacy in a paclitaxel-induced neuropathic pain model. Eur J Med Chem 2025; 290:117510. [PMID: 40101448 DOI: 10.1016/j.ejmech.2025.117510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 03/04/2025] [Accepted: 03/12/2025] [Indexed: 03/20/2025]
Abstract
The severe pain linked to chemotherapy-induced peripheral neuropathy (CIPN) often becomes a critical factor limiting the effective dosage of life-saving chemotherapy treatments. This debilitating side effect not only hampers the effectiveness of cancer therapy but also poses challenges due to the adverse effects of current treatment options for managing CIPN-related pain complications. Soluble epoxide hydrolase (sEH) inhibitors, which elevate endogenous epoxy-fatty acid levels, have been shown to mitigate CIPN-related pain in different rodent models. In our quest to develop potent sEH inhibitors, we developed novel benzyl phenyl urea derivatives incorporating a pyridazinone ring alongside the urea group as a secondary pharmacophore. These compounds demonstrated remarkable potency in inhibiting sEH, with IC50 values ranging from 0.2 to 57 nM. Compound FP9 (IC50 = 0.2 nM), the most potent in this series, exhibited enhanced metabolic stability, translating into significantly improved oral bioavailability (F = 78 %). It consistently relieved pain perception in mice with paclitaxel-induced peripheral neuropathy, achieving a significant and sustained effect compared to gabapentin. In addition, docking studies and molecular dynamics simulations with FP9 provided valuable insights into the binding interactions between the inhibitor and the sEH binding site, offering direction for further optimization of new analogs. These findings align with recent research that highlights the beneficial effects of sEH inhibitors in reducing pain thresholds associated with CIPN.
Collapse
Affiliation(s)
- Deniz Lengerli
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Gazi University, Yenimahalle, 06560, Ankara, Turkey
| | - Arooj Bakht
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Gazi University, Yenimahalle, 06560, Ankara, Turkey
| | - Kübra Çalışkan
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Gazi University, Yenimahalle, 06560, Ankara, Turkey
| | - Philipp Dahlke
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Philosophenweg 14, D-07743, Jena, Germany
| | - Nur Banu Bal
- Department of Pharmacology, Faculty of Pharmacy, Gazi University, Yenimahalle, 06560, Ankara, Turkey
| | - Paul M Jordan
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Philosophenweg 14, D-07743, Jena, Germany
| | - Burcu Çalışkan
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Gazi University, Yenimahalle, 06560, Ankara, Turkey
| | - Oliver Werz
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Philosophenweg 14, D-07743, Jena, Germany
| | - Erden Banoglu
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Gazi University, Yenimahalle, 06560, Ankara, Turkey.
| |
Collapse
|
5
|
Hu W, Bei Y, Chen G, Xu J, Yang M, Yu L, He W, Hu Y, Mao F, Chen S, Xu D, Dai H. Causal insights into the role of metabolites in venous thromboembolism pathogenesis: a metabolome-wide Mendelian randomization study. J Thromb Haemost 2025; 23:1953-1967. [PMID: 40157505 DOI: 10.1016/j.jtha.2025.03.022] [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: 12/02/2024] [Revised: 02/06/2025] [Accepted: 03/21/2025] [Indexed: 04/01/2025]
Abstract
BACKGROUND Venous thromboembolism (VTE) is a significant global health burden, and metabolic alterations play a key role in its pathogenesis. However, previous studies have been constrained by several limitations, hindering clarification of the causal role of metabolites. OBJECTIVES To comprehensively evaluate the causal roles of metabolites in the pathogenesis of VTE and determine whether metabolites mediate the relationships between modifiable risk factors and VTE. METHODS Genetic associations involving 690 plasmas and 211 urinary metabolites were analyzed as exposures, while the outcomes for VTE were derived from a large-scale meta-analysis of genome-wide association studies. Metabolome-wide Mendelian randomization and colocalization analyses were performed to assess the causal role of metabolites in VTE. Metabolic pathway analysis was performed using MetOrigin, and druggability assessments were conducted to prioritize potential therapeutic targets. Additionally, a 2-step Mendelian randomization framework was employed to elucidate the mediating effects of metabolites on the relationships between modifiable risk factors and VTE. RESULTS After Bonferroni correction, 51 plasma metabolites and 18 urinary metabolites were significantly associated with VTE risk. Colocalization evidence supported causal relationships for 37 metabolites with VTE. Eleven metabolic pathways were identified for VTE-related metabolites, and 6 metabolites were prioritized as potential therapeutic targets. Twenty-four modifiable risk factors were associated with 28 VTE-related metabolites, 7 of which were linked to VTE risk. Mediation analyses further revealed significant mediating effect of 8 metabolites on how 6 modifiable factors influenced VTE. CONCLUSION This study identifies potential metabolite biomarkers associated with VTE risk and uncovered the metabolic mediators between modifiable risk factors and VTE, offering new insights for future prevention and treatment strategies.
Collapse
Affiliation(s)
- Wei Hu
- Department of Pharmacy, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yun Bei
- Department of Pharmacy, Affiliated Huzhou Hospital, Zhejiang University School of Medicine, Huzhou, China
| | - Guoquan Chen
- Department of Pharmacy, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Junjun Xu
- Department of Pharmacy, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Mingdong Yang
- Department of Pharmacy, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lingyan Yu
- Department of Pharmacy, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wei He
- Department of Pharmacy, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yani Hu
- Department of Pharmacy, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Fengqian Mao
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Shunan Chen
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Donghang Xu
- Department of Pharmacy, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Haibin Dai
- Department of Pharmacy, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China; Research Center for Clinical Pharmacy, Zhejiang University, Hangzhou, China.
| |
Collapse
|
6
|
Li S, Liu H, Huang W, Yang S, Xie M, Zhou M, Lu B, Li B, Tan B, Yang Y, Dong X. Effect of three polychaetes on growth and reproductive performance, biochemical indices and histology of different tissues in the female Pacific white shrimp, Litopenaeus vannamei broodstock. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2025; 21:49-62. [PMID: 40292183 PMCID: PMC12023780 DOI: 10.1016/j.aninu.2024.11.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 11/14/2024] [Accepted: 11/27/2024] [Indexed: 04/30/2025]
Abstract
A 30-day feeding trial was conducted to assess the effect of three fresh-live polychaetes as diets on growth and reproductive performance, biochemical indices and histology of different tissues in female Pacific white shrimp (Litopenaeus vannamei) broodstock. Two novel polychaete species, Marphysa maxidenticulata (MM group) and Perinereis nuntia (PN group), and a traditional species, Perinereis aibuhitensis (PA group), were used as a single diet and individual experimental groups, respectively. A total of 225 healthy female broodstock shrimp, initial weight of 59.70 ± 0.18 g, were randomly divided into three groups (three replicates of 25 shrimp each). The results showed that the MM group outperformed the PA and PN groups in growth indices, with the highest weight gain, specific growth rate, molting rate, protein efficiency ratio, gonadosomatic index, and lower feed conversion ratio compared to the PN group (P < 0.05). Regarding reproductive performance, the MM group had the highest daily number of sexually mature female broodstock shrimp, successful mating count, maturity rate, mating rate, spawning cycle, total number of fertilized eggs, individual fertilized egg yield, area of mature oocytes, total number of nauplii, naupliar yield/shrimp, hatching rate, and the lowest naupliar deformity rate than the other two groups (P < 0.05). Moreover, compared to the PA and PN groups, the MM group demonstrated superior activities of lipid metabolism-related enzymes and digestive enzymes, and antioxidant capacity in the hepatopancreas, intestine and serum, as well as reduced malondialdehyde levels. Meanwhile, the ovaries of the MM group showed a significant accumulation of triglycerides, estradiol, and vitellogenin compared to the other groups. Histology revealed more developed secretory cells in the hepatopancreas and larger mature oocytes in the MM group compared to the others. In conclusion, M. maxidenticulata can maximize growth, reproductive performance, the activities of lipid metabolism-related enzymes and digestive enzymes, antioxidant and immune ability of female broodstock shrimp. This study demonstrated that M. maxidenticulata could be used as a potential fresh-live diet for the female L. vannamei broodstock.
Collapse
Affiliation(s)
- Shuaipeng Li
- Laboratory of Aquatic Nutrition and Feed, College of Fisheries, Guangdong Ocean University, Zhanjiang, 524088, Guangdong, China
- Guang Dong Engineering Technology Research Center of Aquatic Animals Precision Nutrition and High Efficiency Feed, Zhanjiang, 524088, Guangdong, China
| | - Hao Liu
- Laboratory of Aquatic Nutrition and Feed, College of Fisheries, Guangdong Ocean University, Zhanjiang, 524088, Guangdong, China
- Guang Dong Engineering Technology Research Center of Aquatic Animals Precision Nutrition and High Efficiency Feed, Zhanjiang, 524088, Guangdong, China
| | - Weibin Huang
- Laboratory of Aquatic Nutrition and Feed, College of Fisheries, Guangdong Ocean University, Zhanjiang, 524088, Guangdong, China
- Guang Dong Engineering Technology Research Center of Aquatic Animals Precision Nutrition and High Efficiency Feed, Zhanjiang, 524088, Guangdong, China
| | - Shipei Yang
- Laboratory of Aquatic Nutrition and Feed, College of Fisheries, Guangdong Ocean University, Zhanjiang, 524088, Guangdong, China
- Guang Dong Engineering Technology Research Center of Aquatic Animals Precision Nutrition and High Efficiency Feed, Zhanjiang, 524088, Guangdong, China
| | - Mingsheng Xie
- Laboratory of Aquatic Nutrition and Feed, College of Fisheries, Guangdong Ocean University, Zhanjiang, 524088, Guangdong, China
- Guang Dong Engineering Technology Research Center of Aquatic Animals Precision Nutrition and High Efficiency Feed, Zhanjiang, 524088, Guangdong, China
| | - Menglong Zhou
- Laboratory of Aquatic Nutrition and Feed, College of Fisheries, Guangdong Ocean University, Zhanjiang, 524088, Guangdong, China
- Guang Dong Engineering Technology Research Center of Aquatic Animals Precision Nutrition and High Efficiency Feed, Zhanjiang, 524088, Guangdong, China
| | - Baiquan Lu
- Laboratory of Aquatic Nutrition and Feed, College of Fisheries, Guangdong Ocean University, Zhanjiang, 524088, Guangdong, China
- Guang Dong Engineering Technology Research Center of Aquatic Animals Precision Nutrition and High Efficiency Feed, Zhanjiang, 524088, Guangdong, China
| | - Biao Li
- Laboratory of Aquatic Nutrition and Feed, College of Fisheries, Guangdong Ocean University, Zhanjiang, 524088, Guangdong, China
- Guang Dong Engineering Technology Research Center of Aquatic Animals Precision Nutrition and High Efficiency Feed, Zhanjiang, 524088, Guangdong, China
| | - Beiping Tan
- Laboratory of Aquatic Nutrition and Feed, College of Fisheries, Guangdong Ocean University, Zhanjiang, 524088, Guangdong, China
- Guang Dong Engineering Technology Research Center of Aquatic Animals Precision Nutrition and High Efficiency Feed, Zhanjiang, 524088, Guangdong, China
- Key Laboratory of Aquatic, Livestock and Poultry Feed Science and Technology in South China, Ministry of Agriculture, Zhanjiang, 524000, Guangdong, China
| | - Yuanzhi Yang
- Laboratory of Aquatic Nutrition and Feed, College of Fisheries, Guangdong Ocean University, Zhanjiang, 524088, Guangdong, China
- Guang Dong Engineering Technology Research Center of Aquatic Animals Precision Nutrition and High Efficiency Feed, Zhanjiang, 524088, Guangdong, China
| | - Xiaohui Dong
- Laboratory of Aquatic Nutrition and Feed, College of Fisheries, Guangdong Ocean University, Zhanjiang, 524088, Guangdong, China
- Guang Dong Engineering Technology Research Center of Aquatic Animals Precision Nutrition and High Efficiency Feed, Zhanjiang, 524088, Guangdong, China
- Key Laboratory of Aquatic, Livestock and Poultry Feed Science and Technology in South China, Ministry of Agriculture, Zhanjiang, 524000, Guangdong, China
| |
Collapse
|
7
|
Sarkar R, Sardar SK, Ghosal A, Haldar T, Das K, Ghosh A, Prasad A, Saito-Nakano Y, Dutta S, Nozaki T, Ganguly S. Metronidazole induces prostaglandin E2 formation via arachidonic acid production in protozoan parasite Giardia lamblia. Mol Biochem Parasitol 2025; 262:111676. [PMID: 40101805 DOI: 10.1016/j.molbiopara.2025.111676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 02/14/2025] [Accepted: 03/03/2025] [Indexed: 03/20/2025]
Abstract
The causative agent of giardiasis in human and animals is the amitochondriate Giardia lamblia. We observed that exposing Giardia trophozoites to MTZ led to an increase in lipid peroxidation compared to the control group, which was expressed in terms of menadione production as it is the marker for lipo-peroxidation. Oxidative stress generated by reactive nitrogen species and peroxidation of membrane phospholipids are positively correlated with the enhanced PLA2 activity in several organisms to produce arachidonic acid (AA). Our data suggested Giardia produces a unique 56 kDa dimeric enzyme called Phospholipase B (gPLB) in contrast to higher eukaryotes which was responsible for the production of intracellular free AA. This free AA either reacylates to the cell membrane or deacylates to further produce prostaglandins. In normal un-induced controlled trophozoites the membrane reacylation process was dominant due the higher level of acyle CoA synthase (ACS) expression over the time. However, under the oxidative stressed condition the intracellular ACS expression was down regulated. This led to the increase in deacylation process. When AA deacylation becomes dominant over AA reacylation in cells, the free AA accumulates intracellularly. One of the lipid autacoids, derived from AA is prostaglandin2 (PGE2). Oxidative stress generated by reactive nitrogen species in trophozoites increased the PGE2 production via prostaglandin synthase over the time with respect to the controlled one.
Collapse
Affiliation(s)
- Rituparna Sarkar
- Division of Parasitology, ICMR-National Institute for Research in Bacterial Infections, Kolkata, India
| | - Sanjib Kumar Sardar
- Division of Parasitology, ICMR-National Institute for Research in Bacterial Infections, Kolkata, India
| | - Ajanta Ghosal
- Division of Parasitology, ICMR-National Institute for Research in Bacterial Infections, Kolkata, India
| | - Tapas Haldar
- Division of Parasitology, ICMR-National Institute for Research in Bacterial Infections, Kolkata, India
| | - Koushik Das
- Faculty of Science, Assam down town University, Panikhaiti, Guwahati, Assam 781026, India
| | - Arjun Ghosh
- Department of Biotechnology, Brainware University Barasat, Kolkata, West Bengal, India
| | - Akash Prasad
- Division of Parasitology, ICMR-National Institute for Research in Bacterial Infections, Kolkata, India
| | - Yumiko Saito-Nakano
- Department of Parasitology, National Institute of Infectious Diseases (NIID), Tokyo, Japan
| | - Shanta Dutta
- Division of Bacteriology, ICMR-National Institute for Research in Bacterial Infections, Kolkata, India
| | - Tomoyoshi Nozaki
- Department of Biomedical Chemistry, School of International Health, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Sandipan Ganguly
- Division of Parasitology, ICMR-National Institute for Research in Bacterial Infections, Kolkata, India.
| |
Collapse
|
8
|
Zhang J, Liu L, Lao Z, Lei L, Xu H, Wang X, Lin N, Guo X, Yang J, Tang L. Multi-omics perspective on Huoluo Xiaoling Pellet: key bioactive compounds and related mechanisms for the treatment of knee osteoarthritis. JOURNAL OF ETHNOPHARMACOLOGY 2025; 348:119761. [PMID: 40210178 DOI: 10.1016/j.jep.2025.119761] [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: 10/20/2024] [Revised: 03/31/2025] [Accepted: 04/06/2025] [Indexed: 04/12/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Knee osteoarthritis (KOA) is a prevalent chronic knee joint disease, occurring mainly in the elderly and obese populations. In traditional Chinese medicine (TCM) theory, KOA is categorized as "knee bi" and "bi syndrome," primarily caused by external factors such as wind, cold, and dampness, as well as internal deficiencies. The treatment with Chinese herbal medicine focuses on dispelling wind, cold, and dampness, as well as promoting blood circulation and removing blood stasis. Huoluo Xiaoling Pellet (HXP), derived from Zhang Xichun's "The Records of Traditional Chinese and Western Medicine in Combination " during the Qing Dynasty, was known for its effects of activating blood circulation to remove stasis and alleviating pain by unblocking the collaterals, mainly used for treating conditions with stagnation of qi and blood. It has been included in the "Ancient Classic Prescriptions" by the National Administration of Traditional Chinese Medicine. However, the pharmacodynamic compounds, specific actions, and mechanisms of HXP on KOA have not yet been elucidated. AIM OF STUDY Through qualitative and quantitative analysis, combined with multi-omics technology and network pharmacology, the key bioactive compounds and related mechanisms of HXP in improving knee osteoarthritis were explored. METHODS UHPLC-Q-Orbitrap-MS and HPLC were used to analyze the chemical composition of HXP, chemometric analyses was used for quality control. The therapeutic effect of HXP was evaluated by Anterior Cruciate Ligament Transaction (ACLT)-induced KOA mouse model, and proteomics and lipid metabolomics were combined with network pharmacology and molecular docking techniques to explore for the key bioactive compounds and mechanisms. RESULTS Identified 53 compounds in HXP, with Salvianolic acid B as a key quality marker and potential active compound for KOA. HXP reduced pain, protected chondrocytes, reduced oxidative stress, and improved bone parameters. The combined multi-omics analysis indicates that the protein targets of SHIP1, PTPRC, the cytochrome P450 enzyme family including Cyp1a2, Cyp2c29, Cyp2c50, Cyp2c544, as well as the JAK2/STAT3 pathway, play crucial roles in the improvement of KOA. CONCLUSION Our results demonstrated that HXP was an effective drug to improve the development of KOA. Among them, Salvianolic acid B, the most abundant compound in HXP and the best computer simulation result, may be the key monomer compound of HXP for KOA treatment. The results of the visualisation analysis further contributed to our understanding of HXP treatment of KOA in terms of changes in SHIP1, PTPRC proteins and cytochrome P450 enzyme family. This is associated with the SHIP1/PI3K/AKT and JAK2/STAT3 pathways, as well as arachidonic acid metabolism and immune regulation.
Collapse
Affiliation(s)
- Jingyi Zhang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China; Institute of Traditional Chinese Medicine Intersection and Transformation, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Key Laboratory of Traditional Chinese Medicine Preparations / Guangdong Provincial Technical Engineering Laboratory of Traditional Chinese Medicine Preparations, Guangzhou, 510515, China
| | - Lanbo Liu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China; Institute of Traditional Chinese Medicine Intersection and Transformation, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Key Laboratory of Traditional Chinese Medicine Preparations / Guangdong Provincial Technical Engineering Laboratory of Traditional Chinese Medicine Preparations, Guangzhou, 510515, China
| | - Ziying Lao
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China; Institute of Traditional Chinese Medicine Intersection and Transformation, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Key Laboratory of Traditional Chinese Medicine Preparations / Guangdong Provincial Technical Engineering Laboratory of Traditional Chinese Medicine Preparations, Guangzhou, 510515, China
| | - Li Lei
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China; Institute of Traditional Chinese Medicine Intersection and Transformation, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Key Laboratory of Traditional Chinese Medicine Preparations / Guangdong Provincial Technical Engineering Laboratory of Traditional Chinese Medicine Preparations, Guangzhou, 510515, China
| | - Hui Xu
- Shenzhen Hospital, Beijing University of Chinese Medicine, Shenzhen, 518100, China
| | - Xiwen Wang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China; Institute of Traditional Chinese Medicine Intersection and Transformation, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Key Laboratory of Traditional Chinese Medicine Preparations / Guangdong Provincial Technical Engineering Laboratory of Traditional Chinese Medicine Preparations, Guangzhou, 510515, China
| | - Nianzhen Lin
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China; Institute of Traditional Chinese Medicine Intersection and Transformation, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Key Laboratory of Traditional Chinese Medicine Preparations / Guangdong Provincial Technical Engineering Laboratory of Traditional Chinese Medicine Preparations, Guangzhou, 510515, China
| | - Xiaowen Guo
- Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510315, China.
| | - Jiashun Yang
- The Seventh Affiliated Hospital, Southern Medical University, Foshan, 528244, China.
| | - Ling Tang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China; Institute of Traditional Chinese Medicine Intersection and Transformation, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Key Laboratory of Traditional Chinese Medicine Preparations / Guangdong Provincial Technical Engineering Laboratory of Traditional Chinese Medicine Preparations, Guangzhou, 510515, China; Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510315, China.
| |
Collapse
|
9
|
Gladwell TA, Ferguson D, Clarke N, Brown MD, Gardner P. Insights into the cellular lipid cascade of prostate cells explored using infrared microspectroscopy. Analyst 2025; 150:2280-2287. [PMID: 40326216 DOI: 10.1039/d5an00126a] [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: 05/07/2025]
Abstract
Background: Although prostate cancer (PCa) is the most diagnosed cancer in men worldwide, there is geographical variance in both incidence and morbidity, with higher levels in developed "Western Diet" countries. In particular the high levels of the omega-6 polyunsaturated fatty acid, arachidonic acid (AA), in Western diets has been shown to promote aggressive PCa in vitro. However the exact mechanism through which AA induces the aggressive phenotype has not been fully characterised. Methods: In this study Fourier transform infrared (FTIR) imaging coupled with fluorescence microscopy (FM), is used to follow AA metabolism in PCa cell lines. This is achieved using partially deuterated AA, with a distinctive C-D stretch seen at 2251 cm-1 providing molecular specificity, coupled with Nile Red Fluorescence imaging. Results: We show that, invasive cell lines PC-3, LNCaP C4-2B and DU145 readily uptake and metabolise AA, producing prostaglandins via the COX-2 pathway. Inhibition of the COX-2 pathway with either NS938 or the omega-3 polyunsaturated fatty acid Docosahexaenoic acid (DHA), reduces the invasive stimulus of AA and blocks its uptake. Conclusion: This demonstrates that FTIR imaging can be utilised to follow metabolomics processes within a PCa model and provide an insight to the molecular pathways underlying the cancer metabolome. Additionally, these works provide key insights into the rapid uptake of AA within certain invasive cell lines of prostate cancer, suggesting that AA exposure initiates early cellular responses prior to the uptake and processing of lipids within the cells.
Collapse
Affiliation(s)
- Thomas A Gladwell
- Department of Chemical Engineering, School of Engineering, University of Manchester, Oxford Road, Manchester, M13 9PL, UK.
| | - Dougal Ferguson
- Department of Chemical Engineering, School of Engineering, University of Manchester, Oxford Road, Manchester, M13 9PL, UK.
- Photon Science Institute, University of Manchester, Oxford Road, Manchester, M13 9PL, UK
| | - Noel Clarke
- Department of Surgery, The Christie Hospital NHS Foundation Trust, UK
- Department of Urology, Salford Royal Hospital, UK
| | | | - Peter Gardner
- Department of Chemical Engineering, School of Engineering, University of Manchester, Oxford Road, Manchester, M13 9PL, UK.
- Photon Science Institute, University of Manchester, Oxford Road, Manchester, M13 9PL, UK
| |
Collapse
|
10
|
Wang H, Zhan J, Zhao S, Jiang H, Jia H, Pan Y, Zhong X, Huo J. Sex-induced alterations in rumen microbial communities and metabolite profiles: implications for lamb body weight. BMC Microbiol 2025; 25:328. [PMID: 40426040 PMCID: PMC12107992 DOI: 10.1186/s12866-025-04049-6] [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: 02/28/2025] [Accepted: 05/15/2025] [Indexed: 05/29/2025] Open
Abstract
BACKGROUND Microbiota-metabolome interactions play a crucial role in host physiological regulation and metabolic homeostasis. The aim of this study was to investigate that sex induces alterations in rumen microbial community composition and metabolite profiles in lambs and the influence on body weight. This study aimed to demonstrate that sex- induced alterations in rumen microbial community and metabolite profiles and blood indices and their linkage to growth performance in lambs. RESULTS This study examined (growth indices, serum indices, rumen fermentation parameters, rumen fluid microbiota community and metabolome profiles) in 180 Hu lambs (90 males, and 90 females) with the same age and diet. At six months, male lambs showed significantly greater body weight, serum indices (glutamic pyruvic transaminase, glutamic oxalacetic transaminase, growth hormone, glucagon-like peptide 1, and ghrelin), and molar percentage of propionic acid, isobutyric acid, butyric acid, isovaleric acid and valeric acid compared to female. However, male had lower VFA molar concentrations (acetic acid, propionic acid, butyric acid, and TVFAs), acetic acid/propionic acid, and VFA molar percentage (acetic acid) than female. Significant sex-related differences were observed in rumen microbiota and metabolic enrichment between genders. Moreover, compared with the females lambs, the relative abundance of Succiniclasticum, uncultured_rumen_bacterium, NK4 A214_group, Veillonellaceae_UCG_001 and Butyrivibrio in the male lambs has been significantly increased, while the relative abundance of Prevotella has been significantly decreased (P < 0.05). Notably, there were significant rumen microbiota-metabolite interactions, especially Firmicutes and Bacteroidota as dominant phyla in the sheep rumen with significant differences in correlation with rumen metabolic modules. Additionally, there are pronounced correlations among the microbiota, particularly within the Firmicutes phylum. Furthermore, the up-regulated metabolites in the rumen fluid of male lambs were predominantly enriched in the amino acid metabolite pathway, and these metabolites exhibited a significant positive correlation with body weight. However, the metabolites that were up-regulated in ewe lambs were predominantly enriched in the lipid metabolic pathway, and these metabolites exhibited a significant negative correlation with body weight. Moreover, lamb rumen microbial markers (Lachnospiraceae_UCG_008, Saccharofermentans, unclassified_Clostridia, Christensenellaceae_R_7_group, Anaerovorax, Mogibacterium, and unclassified_Erysipelotrichaceae) and metabolic markers (C75, 4-Coumarate, Flibanserin,3-Amino-5-mercapto-1,2,4-triazole, 1,3-Propane sultone, Fingolimod phosphate ester, S-,) were significantly positively correlated with body weight, but lamb rumen microbial markers (Anaeroplasma, unclassified_Acholeplasmataceae, uncultured_rumen_bacterum_4c28 d_15) and metabolic markers (Mozenavir, Reduced riboflavin, PG(18:2(9Z,12Z)/0:0), Cowanin) were significantly negatively correlated body weight. CONCLUSIONS This study shows that sex-induced alterations in rumen microbial communities and metabolite profiles, adapting to the growth and development of lambs. The findings may help develop targeted strategies to optimize sheep rumen microbiota and improve productivity.
Collapse
Affiliation(s)
- Haibo Wang
- Institute of Animal Husbandry and Veterinary, Jiangxi Academy of Agricultural Science, Nanchang, 330200, China
- Jiangxi Province Key Laboratory of Animal Green and Healthy Breeding, Institute of Animal Husbandry and Veterinary, Jiangxi Academy of Agricultural Science, Nanchang, 330200, China
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, 730070, China
- Provincial Development and Research Institute of Ruminants in Gansu, Lanzhou, 730070, China
| | - Jinshun Zhan
- Institute of Animal Husbandry and Veterinary, Jiangxi Academy of Agricultural Science, Nanchang, 330200, China
- Jiangxi Province Key Laboratory of Animal Green and Healthy Breeding, Institute of Animal Husbandry and Veterinary, Jiangxi Academy of Agricultural Science, Nanchang, 330200, China
| | - Shengguo Zhao
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, 730070, China
- Provincial Development and Research Institute of Ruminants in Gansu, Lanzhou, 730070, China
| | - Haoyun Jiang
- Institute of Animal Husbandry and Veterinary, Jiangxi Academy of Agricultural Science, Nanchang, 330200, China
- Jiangxi Province Key Laboratory of Animal Green and Healthy Breeding, Institute of Animal Husbandry and Veterinary, Jiangxi Academy of Agricultural Science, Nanchang, 330200, China
| | - Haobin Jia
- Institute of Animal Husbandry and Veterinary, Jiangxi Academy of Agricultural Science, Nanchang, 330200, China
- Jiangxi Province Key Laboratory of Animal Green and Healthy Breeding, Institute of Animal Husbandry and Veterinary, Jiangxi Academy of Agricultural Science, Nanchang, 330200, China
| | - Yue Pan
- Institute of Animal Husbandry and Veterinary, Jiangxi Academy of Agricultural Science, Nanchang, 330200, China
- Jiangxi Province Key Laboratory of Animal Green and Healthy Breeding, Institute of Animal Husbandry and Veterinary, Jiangxi Academy of Agricultural Science, Nanchang, 330200, China
- College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin, 300384, China
| | - Xiaojun Zhong
- Institute of Animal Husbandry and Veterinary, Jiangxi Academy of Agricultural Science, Nanchang, 330200, China
- Jiangxi Province Key Laboratory of Animal Green and Healthy Breeding, Institute of Animal Husbandry and Veterinary, Jiangxi Academy of Agricultural Science, Nanchang, 330200, China
| | - Junhong Huo
- Institute of Animal Husbandry and Veterinary, Jiangxi Academy of Agricultural Science, Nanchang, 330200, China.
- Jiangxi Province Key Laboratory of Animal Green and Healthy Breeding, Institute of Animal Husbandry and Veterinary, Jiangxi Academy of Agricultural Science, Nanchang, 330200, China.
| |
Collapse
|
11
|
Sala-Climent M, Lal E, Cedola F, Alharthi M, Fernandez-Bustamante M, Agustin-Perez M, Singh A, Choi SI, Rivera T, Nguyen K, Lee S, Golshan S, Holt T, Quehenberger O, Coras R, Guma M. Oxylipin serum profile changes in response to an open-label anti-inflammatory dietary intervention. Clin Nutr ESPEN 2025; 68:389-402. [PMID: 40414498 DOI: 10.1016/j.clnesp.2025.05.027] [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: 11/15/2024] [Revised: 05/13/2025] [Accepted: 05/16/2025] [Indexed: 05/27/2025]
Abstract
INTRODUCTION Oxylipins are bioactive lipids involved in inflammation. This study evaluated how a 2-week anti-inflammatory diet (ITIS, omega-3/omega-6 ratio of 1:1.5) affects plasma oxylipin profiles in patients with active Rheumatoid Arthritis (RA). METHODS In an open-label pilot trial, 20 RA patients (≥3 tender and ≥3 swollen joints) followed the ITIS diet. Targeted lipidomics by mass spectrometry was used to quantify oxylipins. Patients were classified as responders or non-responders based on ≥50 % pain reduction (Pain-50). Dietary intake was assessed through diet scores, and statistical analyses were performed using RStudio. RESULTS Participants were predominantly female (90 %) with an average age of 57.1. At baseline, responders consumed more walnuts (p = 0.08), almond milk (p = 0.06), avocado (p = 0.04), and quinoa (p = 0.05), and fewer burgers (p = 0.02). No differences in diet adherence were observed between groups. Baseline oxylipin levels did not differ significantly. However, after the intervention, six oxylipins-5-HETE, 11,12-diHETrE, 14,15-diHETrE, 19,20-DiHDPA, 9-oxo-ODE, and 14,15-EET-differed significantly between responders and non-responders. Notably, oxylipins derived from both arachidonic acid (omega-6) and eicosapentaenoic acid (omega-3) decreased significantly after the diet (p = 0.0006 and p = 0.01, respectively). CONCLUSION The anti-inflammatory diet modified circulating levels of both pro- and anti-inflammatory oxylipins. These changes varied by pain response, suggesting that diet can influence inflammatory pathways in RA. Further studies are warranted to clarify the mechanisms linking dietary changes, oxylipin modulation, and clinical outcomes. CLINICAL TRIALS IDENTIFIER NCT04999683.
Collapse
Affiliation(s)
- Marta Sala-Climent
- Department of Medicine, School of Medicine, University of California, 9500 Gilman Drive, San Diego, CA 92093, USA
| | - Esha Lal
- Department of Medicine, School of Medicine, University of California, 9500 Gilman Drive, San Diego, CA 92093, USA
| | - Francesca Cedola
- Department of Medicine, School of Medicine, University of California, 9500 Gilman Drive, San Diego, CA 92093, USA
| | - Maram Alharthi
- Department of Medicine, School of Medicine, University of California, 9500 Gilman Drive, San Diego, CA 92093, USA
| | - Marta Fernandez-Bustamante
- Department of Medicine, School of Medicine, University of California, 9500 Gilman Drive, San Diego, CA 92093, USA
| | - Meritxell Agustin-Perez
- Department of Medicine, School of Medicine, University of California, 9500 Gilman Drive, San Diego, CA 92093, USA
| | - Abha Singh
- Department of Medicine, School of Medicine, University of California, 9500 Gilman Drive, San Diego, CA 92093, USA
| | - Soo-In Choi
- Department of Medicine, School of Medicine, University of California, 9500 Gilman Drive, San Diego, CA 92093, USA
| | - Tania Rivera
- Department of Medicine, School of Medicine, University of California, 9500 Gilman Drive, San Diego, CA 92093, USA
| | - Katherine Nguyen
- Department of Medicine, School of Medicine, University of California, 9500 Gilman Drive, San Diego, CA 92093, USA
| | - Susan Lee
- Department of Medicine, School of Medicine, University of California, 9500 Gilman Drive, San Diego, CA 92093, USA
| | - Shahrokh Golshan
- Psychiatry, School of Medicine, University of California, 9500 Gilman Drive, San Diego, CA 92093, USA
| | - Tiffany Holt
- Psychiatry, School of Medicine, University of California, 9500 Gilman Drive, San Diego, CA 92093, USA
| | - Oswald Quehenberger
- Pharmacology, School of Medicine, University of California, 9500 Gilman Drive, San Diego, CA 92093, USA
| | - Roxana Coras
- Department of Medicine, School of Medicine, University of California, 9500 Gilman Drive, San Diego, CA 92093, USA
| | - Monica Guma
- Department of Medicine, School of Medicine, University of California, 9500 Gilman Drive, San Diego, CA 92093, USA; VA San Diego Healthcare System, 3350 La Jolla Village Dr, San Diego, CA 92161, USA.
| |
Collapse
|
12
|
Bonglack EN, Hill KK, Barry AP, Bartlett A, Castellano-Escuder P, Hirschey MD, Luftig MA. Fatty acid desaturases link cell metabolism pathways to promote proliferation of Epstein-Barr virus-infected B cells. PLoS Pathog 2025; 21:e1012685. [PMID: 40403013 DOI: 10.1371/journal.ppat.1012685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Accepted: 04/24/2025] [Indexed: 05/24/2025] Open
Abstract
Epstein-Barr virus (EBV) is a gamma herpesvirus that infects up to 95% of the human population by adulthood, typically remaining latent in the host memory B cell pool. In immunocompromised individuals, EBV can drive the transformation and rapid proliferation of infected B cells, ultimately resulting in neoplasia. The same transformation process can be induced in vitro, with EBV-infected peripheral blood B cells forming immortalized lymphoblastoid cell lines (LCLs) within weeks. In this study, we found that the fatty acid desaturases stearoyl-CoA desaturase 1 (SCD1) and fatty acid desaturase 2 (FADS2) are upregulated by EBV and crucial for EBV-induced B cell proliferation. We show that pharmacological and genetic inhibition of both SCD1 and FADS2 results in a significantly greater reduction in proliferation and cell cycle arrest, compared to perturbing either enzyme individually. Additionally, we found that inhibiting either SCD1 or FADS2 alone hypersensitizes LCLs to palmitate-induced apoptosis. Further free fatty acid profiling and metabolic analysis of dual SCD1/FADS2-inhibited LCLs revealed an increase in free unsaturated fatty acids, a reduction of oxidative phosphorylation, and a reduction of glycolysis, thereby linking the activity of SCD1 and FADS2 to overall growth-promoting metabolism. Lastly, we show that SCD1 and FADS2 are important in the growth of clinically derived EBV+ immunoblastic lymphoma cells. Collectively, these data demonstrate a previously uncharacterized role of lipid desaturation in EBV+ transformed B cell proliferation, revealing a metabolic pathway that can be targeted in future anti-lymphoma therapies.
Collapse
Affiliation(s)
- Emmanuela N Bonglack
- Department of Molecular Genetics and Microbiology, Center for Virology, Duke University School of Medicine, Durham, North Carolina, United States of America
- Cardiovascular Epidemiology Unit, University of Cambridge, Cambridge, United Kingdom
| | - Kaeden K Hill
- Department of Molecular Genetics and Microbiology, Center for Virology, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Ashley P Barry
- Department of Molecular Genetics and Microbiology, Center for Virology, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Alexandria Bartlett
- Department of Molecular Genetics and Microbiology, Center for Virology, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Pol Castellano-Escuder
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Matthew D Hirschey
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Micah A Luftig
- Department of Molecular Genetics and Microbiology, Center for Virology, Duke University School of Medicine, Durham, North Carolina, United States of America
| |
Collapse
|
13
|
Hu Q, Zhang J, Luo X, Hu P, Li J, Li F, Wang Z, Zhang S, Jiao Z, Liu Y, Duanmu J, Jin L, Xie P, Zhu W, Zheng W, Shang H, Hu X, Chen Z, Xiao RP, Zhang Y. Intracellular L-PGDS-Derived 15d-PGJ2 Inhibits CaMKII Through Lipoxidation to Alleviate Cardiac Ischemia/Reperfusion Injury. Circulation 2025. [PMID: 40396239 DOI: 10.1161/circulationaha.124.070936] [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: 08/19/2024] [Accepted: 04/09/2025] [Indexed: 05/22/2025]
Abstract
BACKGROUND Myocardial ischemia/reperfusion (I/R) injury is a substantial challenge to the management of ischemic heart disease, the leading cause of mortality worldwide. Arachidonic acid (AA) is a prominent polyunsaturated fatty acid in the human body and plays an important role in various physiological and pathological conditions. AA metabolic enzymes determine AA levels; however, currently there is no comprehensive analysis of AA enzymes in cardiac I/R injury. METHODS The profiling of AA metabolic enzymes was analyzed with the RNA sequencing transcriptome data from the mouse heart tissues with I/R injury. Cultured neonatal and adult rat ventricular myocytes, human embryonic stem cell-derived cardiomyocytes, and in vivo mouse I/R models were used to confirm the role of L-PGDS (lipocalin-type prostaglandin D2 synthase)/15d-PGJ2 in I/R injury. A biotin-tagged 15d-PGJ2 analog combined with liquid chromatography-tandem mass spectrometry was used to identify the downstream signaling of L-PGDS/15d-PGJ2. RESULTS Based on the transcriptome data and experimental validations, L-PGDS, together with its downstream metabolite 15d-PGJ2, was downregulated in cardiac tissue with I/R injury. Functionally, L-PGDS overexpression mitigates myocardial I/R injury, whereas knockdown exacerbates the damage. Supplementation of 15d-PGJ2 alleviated I/R injury. Mechanistically, 15d-PGJ2 covalently bound to the Ca2+/CaMKII (calmodulin protein kinase II) and induced lipoxidation of its cysteine 495 (CaMKII-δ9) to dampen the formation of CaMKII oligomers and alleviate its overactivation, consequently ameliorating cardiomyocyte death and cardiac injury. CONCLUSIONS Our study uncovered L-PGDS/15d-PGJ2/CaMKII signaling as a new mechanism underlying I/R-induced cardiomyocyte death. This provides new mechanistic insights and therapeutic targets for myocardial I/R injury and subsequent heart failure. We also showed that lipoxidation is a new post-translational modification type for CaMKII, deepening our understanding of the regulation of its activity.
Collapse
Affiliation(s)
- Qingmei Hu
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, College of Future Technology, Peking University, China. (Q.H., J.L., F.L., L.J., P.X., W. Zheng, H.S., X.H., Z.C., R.-P.X.)
| | - Junxia Zhang
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, China. (J.Z.)
- Beijing Key Laboratory of Cardiovascular Receptors Research, China (J.Z., Y.Z.)
- Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Haihe Laboratory of Cell Ecosystem, Beijing, China (J.Z.)
| | - Xile Luo
- Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University Health Science Center, Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, China. (X.L., X.D., Y.Z.)
| | - Peiyu Hu
- Institute of Energy, Peking University, China. (P.H.)
| | - Jiayi Li
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, College of Future Technology, Peking University, China. (Q.H., J.L., F.L., L.J., P.X., W. Zheng, H.S., X.H., Z.C., R.-P.X.)
| | - Fan Li
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, College of Future Technology, Peking University, China. (Q.H., J.L., F.L., L.J., P.X., W. Zheng, H.S., X.H., Z.C., R.-P.X.)
| | - Zeyuan Wang
- Department of Cardiology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China. (Z.W., S.Z.)
| | - Shuyang Zhang
- Department of Cardiology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China. (Z.W., S.Z.)
| | - Zishan Jiao
- Institute of Basic Medical Sciences & School of Basic Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China. (Z.J.)
| | - Yitong Liu
- Peking-Tsinghua Center for Life Sciences, Peking University, China. (Y.L., W. Zhu, Z.C., R.-P.X.)
| | | | - Li Jin
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, College of Future Technology, Peking University, China. (Q.H., J.L., F.L., L.J., P.X., W. Zheng, H.S., X.H., Z.C., R.-P.X.)
| | - Peng Xie
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, College of Future Technology, Peking University, China. (Q.H., J.L., F.L., L.J., P.X., W. Zheng, H.S., X.H., Z.C., R.-P.X.)
| | - Wenneng Zhu
- Peking-Tsinghua Center for Life Sciences, Peking University, China. (Y.L., W. Zhu, Z.C., R.-P.X.)
- Department of Chemical Biology, College of Chemistry, Peking University, China. (W. Zhu)
| | - Wen Zheng
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, College of Future Technology, Peking University, China. (Q.H., J.L., F.L., L.J., P.X., W. Zheng, H.S., X.H., Z.C., R.-P.X.)
| | - Haibao Shang
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, College of Future Technology, Peking University, China. (Q.H., J.L., F.L., L.J., P.X., W. Zheng, H.S., X.H., Z.C., R.-P.X.)
| | - Xinli Hu
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, College of Future Technology, Peking University, China. (Q.H., J.L., F.L., L.J., P.X., W. Zheng, H.S., X.H., Z.C., R.-P.X.)
| | - Zhixing Chen
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, College of Future Technology, Peking University, China. (Q.H., J.L., F.L., L.J., P.X., W. Zheng, H.S., X.H., Z.C., R.-P.X.)
- Peking-Tsinghua Center for Life Sciences, Peking University, China. (Y.L., W. Zhu, Z.C., R.-P.X.)
| | - Rui-Ping Xiao
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, College of Future Technology, Peking University, China. (Q.H., J.L., F.L., L.J., P.X., W. Zheng, H.S., X.H., Z.C., R.-P.X.)
- Peking-Tsinghua Center for Life Sciences, Peking University, China. (Y.L., W. Zhu, Z.C., R.-P.X.)
- Beijing City Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, China. (R.-P.X.)
- PKU-Nanjing Joint Institute of Translational Medicine, China (R.-P.X.)
| | - Yan Zhang
- Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University Health Science Center, Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, China. (X.L., X.D., Y.Z.)
- Beijing Key Laboratory of Cardiovascular Receptors Research, China (J.Z., Y.Z.)
- NHC Key Laboratory of Cell Transplantation, First Affiliated Hospital of Harbin Medical University, China (Y.Z.)
- Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, China (Y.Z.)
| |
Collapse
|
14
|
Chang J, Zhang L, An Q, Ma Z, Xu P, Cernava T, Jin D. Novel insight into the mechanisms of neurotoxicity induced by type I and type II pyrethroids via disrupting the gut-brain axis in lizards. THE SCIENCE OF THE TOTAL ENVIRONMENT 2025; 983:179697. [PMID: 40398162 DOI: 10.1016/j.scitotenv.2025.179697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2025] [Revised: 05/13/2025] [Accepted: 05/15/2025] [Indexed: 05/23/2025]
Abstract
Type I and type II pyrethroids are widely used and frequently detected in agricultural environments. The neurotoxic effects and underlying mechanisms of pyrethroids in native animal populations, including lizards as common farmland inhabitants, remain unclear. This study exposed male lizards (Eremias argus) to type I bifenthrin (BF) and type II fluvalinate (FA) pyrethroids for 28 days, resulting in abnormal behaviors. Targeted analyses indicated that neurotransmitters, including dopamine, GABA, acetylcholine, and choline in lizard plasma, were significantly decreased with alterations in the cholinergic synapse, dopaminergic synapse, and cAMP signaling pathway in the brain after BF and FA treatment. Nervous system-related genes such as CACNA1A, CACNA1B, and CACNA1C were significantly down-regulated and highly correlated with arachidonic acid metabolism pathway-related metabolites in lizard gut. A notable decrease in metabolites within the arachidonic acid metabolism pathway and alterations in the gut microbiome were indicative for anti-inflammatory responses and neurotoxic effects. Interestingly, increased type I BF bioaccumulation in lizard intestines induced a higher abundance of Akkermansia, which resulted in reduced inflammation in the gut and lower neurotoxic effects compared to the low-dose BF exposure group. This study reveals contrasting dose-responses between pyrethroid types and suggests gut-brain axis-regulated neurotoxicity in lizards.
Collapse
Affiliation(s)
- Jing Chang
- Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Leisen Zhang
- Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qiong An
- Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zheng Ma
- Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Peng Xu
- Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Tomislav Cernava
- Institute of Environmental Biotechnology, Graz University of Technology, Petersgasse 12, 8010 Graz, Austria; School of Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton SO17 1BJ, United Kingdom
| | - Decai Jin
- Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China.
| |
Collapse
|
15
|
Cui Y, Li Z, Lai M, Yang Y, Zhang Z, Feng Y, Yao M, Li J. Therapeutic effects of Saussurea graminea Dunn and its active compounds in sepsis-associated liver injury: Transcriptomics, metabolomics and experimental validation. JOURNAL OF ETHNOPHARMACOLOGY 2025; 349:119985. [PMID: 40383246 DOI: 10.1016/j.jep.2025.119985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2025] [Revised: 05/04/2025] [Accepted: 05/14/2025] [Indexed: 05/20/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Saussurea graminea Dunn (SG), a traditional Chinese medicinal herb known as "Za Chi" in Tibet of China, is frequently utilized in the treatment of inflammatory diseases such as hepatitis. However, the active ingredients and mechanism of its therapeutic effect on Sepsis - associated liver injury (SALI) remain unclear. AIM OF THE STUDY To elucidate the effect of SG in combating SALI, uncover its mechanism of action, and explore possible active compounds. MATERIALS AND METHODS We established a SALI model by intraperitoneal injection of lipopolysaccharide to assess the efficacy of SG. Transcriptomics and metabolomics were employed to reveal its possible mechanism of action. Subsequently, Western blot, flow cytometry, confocal microscopy, quantitative PCR, HPLC-MS, and molecular docking were utilized to verify its mechanism and active ingredients. RESULTS SG effectively counteracts SALI by inhibiting the cytokine storm. Transcriptomics indicates that SG regulates SALI through mitochondrial/TNF and metabolic pathways. Metabolomics demonstrates that arachidonic acid metabolism is involved in the process of SG treating SALI. HPLC-MS identified the main components of SG as chlorogenic acid, syringin, scopoletin, rutin, isochlorogenic acid, and narcissin, and these six compounds were confirmed as potential active components in the RAW264.7 inflammation model. CONCLUSION SG and its active ingredients play a role in alleviating SALI by reducing the cytokine storm through mtDNA/TNF/arachidonic acid metabolism.
Collapse
Affiliation(s)
- Yushun Cui
- The National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330006, China
| | - Zhiqiang Li
- The National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330006, China
| | - Miao Lai
- The National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330006, China
| | - Ying Yang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Zhengwen Zhang
- The National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330006, China
| | - Yulin Feng
- The National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330006, China.
| | - Min Yao
- The National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330006, China.
| | - Junmao Li
- The National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330006, China.
| |
Collapse
|
16
|
Balck A, Borsche M, Campbell P, Luo X, Harvey J, Brückmann T, Ludwig C, Harms A, Lohmann K, Brown E, Morris HR, Schapira AH, Hankemeier T, Fleming R, Szymczak S, Klein C. The role of dopaminergic medication and specific pathway alterations in idiopathic and PRKN/PINK1-mediated Parkinson's disease. SCIENCE ADVANCES 2025; 11:eadp7063. [PMID: 40367158 PMCID: PMC12077494 DOI: 10.1126/sciadv.adp7063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 04/09/2025] [Indexed: 05/16/2025]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease, with a rapidly increasing prevalence worldwide. Biomarkers monitoring state and progression are urgently needed, and metabolomics from easily accessible biofluids holds the potential to elucidate pathophysiological underpinnings in PD. Several studies suggested metabolomic differences between patients and controls, but findings are controversial, and independent replication is scarce. We thus applied state-of-the-art, large-scale metabolomics in patients with idiopathic and monogenic PD and controls from two independent samples, analyzed by a strict meta-analysis approach. Thereby, we (i) debunked that l-Dopa medication and not disease status causes the most substantial metabolomic differences and (ii) identified polyamine metabolism alterations, partly, but not entirely associated with l-Dopa treatment. Furthermore, we found explorative but robust evidence for alterations in endocannabinoid metabolites; detected lipid metabolism alterations, highlighting potential crosslinks with alpha-synuclein pathology; and provided evidence for a metabolomic signature for the role of oxidative damage in patients with PRKN- and PINK1-linked PD.
Collapse
Affiliation(s)
- Alexander Balck
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
- Department of Neurology, University of Lübeck and University Hospital Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Max Borsche
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
- Department of Neurology, University of Lübeck and University Hospital Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Philip Campbell
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK
| | - Xi Luo
- Digital Twin Center, School of Medicine, University of Galway, University Rd, Galway, Ireland
| | - John Harvey
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK
| | | | - Charlotte Ludwig
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | - Amy Harms
- Digital Twin Center, School of Medicine, University of Galway, University Rd, Galway, Ireland
| | - Katja Lohmann
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | - Emmeline Brown
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK
| | - Huw R. Morris
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK
| | - Anthony H. Schapira
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK
| | - Thomas Hankemeier
- Digital Twin Center, School of Medicine, University of Galway, University Rd, Galway, Ireland
| | - Ronan Fleming
- Digital Twin Center, School of Medicine, University of Galway, University Rd, Galway, Ireland
- Metabolomics and Analytics Centre, Leiden Academic Centre for Drug Research, Leiden University, Leiden, Netherlands
| | - Silke Szymczak
- Institute of Medical Biometry and Statistics, University of Lübeck, Lübeck, Germany
| | - Christine Klein
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| |
Collapse
|
17
|
Zheng PC, Pan XQ, Zhou YJ, Lai KP, Li R, Zhang XX. Unraveling the impact of micro- and nano-sized polymethyl methacrylate on gut microbiota and liver lipid metabolism: Insights from oral exposure studies. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 373:126157. [PMID: 40157484 DOI: 10.1016/j.envpol.2025.126157] [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: 12/10/2024] [Revised: 02/14/2025] [Accepted: 03/27/2025] [Indexed: 04/01/2025]
Abstract
Microplastics, particularly polymethyl methacrylate (PMMA), have emerged as significant environmental pollutants, with growing concerns about their impact on various biological processes. However, the effects of chronic PMMA exposure on hepatic lipid metabolism remain insufficiently studied. This research aimed to examine the consequences of chronic exposure to PMMA particles of different sizes (100 nm and 2 μm) on hepatic lipid metabolism in mice. Female C57BL/6J mice were administered PMMA particles in drinking water over an 8-week period, and the effects on intestinal and liver morphology and function were evaluated. Histopathological analyses, gut microbiota profiling, and serum and liver assays were conducted to assess oxidative stress, lipid metabolism-related biomarkers, and liver metabolomics. The results revealed that PMMA particles accumulated in both the liver and colon, causing liver injury characterized by elevated ALT and AST levels. The exposure also induced oxidative stress by inhibiting the NRF2/HO-1 signaling pathway. Furthermore, PMMA exposure resulted in significant alterations to the gut microbiota and hepatic metabolism. These changes were linked to increased microbial diversity, which impacted cholesterol metabolism through the gut-liver axis. Additionally, the activation of the PI3K/AKT/PPARγ signaling pathway disrupted hepatic lipid metabolism, leading to increased cholesterol synthesis and hepatic lipid accumulation. This study underscores the potential of PMMA to disrupt both hepatic lipid metabolism and gut microbiota composition, suggesting a novel mechanism by which PMMA exposure could contribute to metabolic disorders and liver disease.
Collapse
Affiliation(s)
- Peng Chen Zheng
- College of Basic Medical Sciences, Guilin Medical University, Guilin, 541100, China
| | - Xin Qiang Pan
- College of Basic Medical Sciences, Guilin Medical University, Guilin, 541100, China
| | - Yi Jiong Zhou
- College of Basic Medical Sciences, Guilin Medical University, Guilin, 541100, China
| | - Keng Po Lai
- Key Laboratory of Environmental Pollution and Integrative Omics, Education Department of Guangxi Zhuang Autonomous Region, Guilin Medical University, Guilin, 541100, China; Department of Applied Science, Hong Kong Metropolitan University, Hong Kong, China
| | - Rong Li
- College of Basic Medical Sciences, Guilin Medical University, Guilin, 541100, China; Key Laboratory of Environmental Pollution and Integrative Omics, Education Department of Guangxi Zhuang Autonomous Region, Guilin Medical University, Guilin, 541100, China.
| | - Xiao Xi Zhang
- Key Laboratory of Environmental Pollution and Integrative Omics, Education Department of Guangxi Zhuang Autonomous Region, Guilin Medical University, Guilin, 541100, China; Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin, 541100, China.
| |
Collapse
|
18
|
Zhang ZY, Wang YM, Wang N, Wang YS, Zhang H, Wang D, Wang LX, Cui HT, Wen WB, Lv SQ, Cao YJ. Shenzhuo formulation ameliorates diabetic nephropathy by regulating cytochrome P450-mediated arachidonic acid metabolism. World J Diabetes 2025; 16:103511. [DOI: 10.4239/wjd.v16.i5.103511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 12/31/2024] [Accepted: 02/17/2025] [Indexed: 04/25/2025] Open
Abstract
BACKGROUND Diabetic nephropathy (DN) is a major complication of diabetes, marked by progressive renal damage and an inflammatory response. Although research has investigated the pathological mechanisms underlying DN, effective treatment options remain limited.
AIM To evaluate the therapeutic impact of Shenzhuo formulation (SZF) on a DN mouse model and to examine its potential molecular mechanisms using transcriptomic and metabolomic approaches.
METHODS We established a DN mouse model through a high-fat diet combined with streptozotocin (STZ) injection, followed by SZF treatment. We analyzed SZF’s effects on gene expression and metabolite profiles in renal tissues of DN mice using transcriptomics and metabolomics techniques. Additionally, based on transcriptomic and non-targeted metabolomic findings, we further assessed SZF’s influence on the expression of factors related to the cytochrome P450 (CYP450)-mediated arachidonic acid (AA) metabolism pathway, as well as its effects on inflammation and oxidative stress.
RESULTS SZF intervention significantly decreased hyperglycemia and mitigated renal function impairment in DN mice. Pathological analysis revealed that SZF treatment improved renal tissue damage, reduced fibrosis, and diminished glycogen deposition. Transcriptomic analysis indicated that SZF influenced mRNA expression of CYP450-related genes, including Cyp2j13, Cyp2b9, Pla2 g2e/Cyp4a12a, Cyp4a32, Cyp2e1, and Cyp4a14. Non-targeted metabolomic results demonstrated that SZF altered the levels of metabolites associated with the AA metabolic pathway, including 5,6-EET, 14,15-EET, phosphatidylcholine, and 20-HETE. Further experiments showed that SZF upregulated the expression of CYP4A and CYP2E proteins in renal tissue, as well as CYP2J and CYP2B proteins. Additionally, SZF significantly reduced the expression of inflammatory factors in renal tissue, enhanced antioxidant enzyme activity, and alleviated oxidative stress.
CONCLUSION SZF exerts anti-inflammatory and antioxidant effects by regulating CYP450-mediated AA metabolism, leading to improved renal function and improved pathological state in DN mice.
Collapse
Affiliation(s)
- Zhong-Yong Zhang
- Department of Endocrinology, Cangzhou Hospital of Integrated Traditional Chinese Medicine and Western Medicine of Hebei Province Affiliated to Hebei University of Chinese Medicine, Cangzhou 061012, Hebei Province, China
| | - Yu-Ming Wang
- College of Integrative Chinese and Western Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Ning Wang
- The First School of Clinical Medicine, Yunnan University of Chinese Medicine, Kunming 650500, Yunnan Province, China
| | - Yuan-Song Wang
- Department of Endocrinology, Cangzhou Hospital of Integrated Traditional Chinese Medicine and Western Medicine of Hebei Province Affiliated to Hebei University of Chinese Medicine, Cangzhou 061012, Hebei Province, China
| | - Hui Zhang
- Department of Endocrinology, Cangzhou Hospital of Integrated Traditional Chinese Medicine and Western Medicine of Hebei Province Affiliated to Hebei University of Chinese Medicine, Cangzhou 061012, Hebei Province, China
| | - Duo Wang
- North China University of Science and Technology, Tangshan 063000, Hebei Province, China
| | - Li-Xin Wang
- Department of Endocrinology, Cangzhou Hospital of Integrated Traditional Chinese Medicine and Western Medicine of Hebei Province Affiliated to Hebei University of Chinese Medicine, Cangzhou 061012, Hebei Province, China
| | - Huan-Tian Cui
- The First School of Clinical Medicine, Yunnan University of Chinese Medicine, Kunming 650500, Yunnan Province, China
| | - Wei-Bo Wen
- The First School of Clinical Medicine, Yunnan University of Chinese Medicine, Kunming 650500, Yunnan Province, China
| | - Shu-Quan Lv
- Department of Endocrinology, Cangzhou Hospital of Integrated Traditional Chinese Medicine and Western Medicine of Hebei Province Affiliated to Hebei University of Chinese Medicine, Cangzhou 061012, Hebei Province, China
| | - Yong-Jun Cao
- Department of Endocrinology, Nantong Affiliated Hospital, Nanjing University of Traditional Chinese Medicine, Nantong 226000, Jiangsu Province, China
| |
Collapse
|
19
|
Li L, Sun Z, Sun W, Zhai Y, Ding N, Wang W. Associations between ionomic profile and metabolic abnormalities in a murine model of sodium sulfide induced alopecia areata. Front Pharmacol 2025; 16:1507348. [PMID: 40438604 PMCID: PMC12117264 DOI: 10.3389/fphar.2025.1507348] [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: 10/07/2024] [Accepted: 04/28/2025] [Indexed: 06/01/2025] Open
Abstract
Background Alopecia areata (AA) is a common autoimmune disorder marked by non-scarring hair loss, which imposes significant psychosocial stress on patients. To investigate key metabolites and ions involved in AA's pathogenesis, we utilized gas chromatography-mass spectrometry (GC-MS) for non-targeted metabolomics and inductively coupled plasma mass spectrometry (ICP-MS) for ionomics. Methods A total of 36 six-week-old Kunming mice were divided into control (n = 12), an AA model (n = 12), and tofacitinib-treated groups (n = 12). A mouse model of AA was established by sodium sulfide (Na2S) induction in both the model and treatment groups, while the treatment group (n = 12) received tofacitinib treatment at a dose of 1 mg/kg. GC-MS was used to determine the metabolic profiling in serum samples, and ICP-MS was applied to assess ionomic changes in the serum samples. Potential metabolites and ions were identified using orthogonal partial least squares-discriminant analysis (OPLS-DA). Subsequently, MetaboAnalyst 5.0 and the Kyoto Encyclopedia of Genes and Genomes database (KEGG) were used to map the metabolic pathways. Spearman correlation analysis was conducted to identify relationships and potential regulatory interactions between differential metabolites and individual ions. Results Metabolomics analysis revealed that D-lactic acid, glycolic acid, linoleic acid, petroselinic acid, and stearic acid are key differential metabolites between the control, AA model, and tofacitinib groups. Pathway analysis highlighted that the biosynthesis of unsaturated fatty acids and linoleic acid metabolism are pivotal pathways implicated in the onset and progression of AA. Furthermore, ionomics analysis identified magnesium, aluminum, titanium, and nickel as differential ions among the three groups. The integrated metabolomics and ionomics analysis indicated that linoleic acid, a key differential metabolite according to the KEGG database, shows a positive correlation with phosphorus, vanadium, magnesium, and zinc. Among these, Mg2+ (Mg2+) play a crucial role in modulating CD8+ T cell infiltration, thereby influencing the disease progression in AA. Conclusion Tofacitinib inhibits CD8+ T cell infiltration in hair follicles affected by sodium sulfide-induced AA by modulating the linoleic acid metabolism-Mg2+ pathway. Our findings offer new insights and potential avenues for the clinical diagnosis and treatment of AA, suggesting that targeting metabolic and ionic pathways could enhance therapeutic outcomes.
Collapse
Affiliation(s)
- Luning Li
- Clinical Medical Laboratory Center, Jining First People’s Hospital, Jining, Shandong, China
| | - Zhen Sun
- Department of Clinical Pharmacy, Jining First People’s Hospital, Jining, Shandong, China
| | - Wenxue Sun
- Department of Clinical & Translational Medicine, Jining First People’s Hospital, Jining, Shandong, China
| | - Yujuan Zhai
- Department of Dermatology, Jining First People’s Hospital, Jining, Shandong, China
| | - Na Ding
- Department of Dermatology, Jining First People’s Hospital, Jining, Shandong, China
| | - Wei Wang
- Department of Dermatology, Jining First People’s Hospital, Jining, Shandong, China
| |
Collapse
|
20
|
You H, Liang Y. Atopic Dermatitis: The Relationship Between Immune Mediators and Skin Lipid Barrier. Clin Rev Allergy Immunol 2025; 68:49. [PMID: 40366491 DOI: 10.1007/s12016-025-09057-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/24/2025] [Indexed: 05/15/2025]
Abstract
Atopic dermatitis (AD) is a chronic inflammatory skin disease that is prevalent worldwide with complex etiology. Skin barrier defects and abnormal immune activation are crucial in the occurrence and development of AD. In the classic model of the skin barrier, lipids are essential for the formation and maintenance of this barrier as a "mortar" component. However, abnormally activated immune responses promote the lipid barrier deficiency through the secretion of various types of immune mediators directly or indirectly. In this review, we first introduce the skin lipid barrier (SLB) under both normal and abnormal conditions, highlighting the contributions of lipids derived from keratinocytes and sebaceous glands (SGs). Subsequently, the relationships between the immune mediators of Th1, Th2, Th17, Th22, and other types (adipokines, prostaglandins, leukotrienes) and SLB are elaborated in turn. Finally, the therapies for restoring SLB to treat AD are summarized, with a focus on the restoration effect of dupilumab on SLB. We hope that this review will offer a comprehensive perspective for understanding the pathogenesis of lipid metabolism disorders and SLB deficiency caused by immune mediators in AD. It also aims to provide guidance for further research on targeting inflammatory mediators to restore SLB.
Collapse
Affiliation(s)
- Huayan You
- Hunan Key Laboratory of Medical Epigenomics & Department of Dermatology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Yunsheng Liang
- Hunan Key Laboratory of Medical Epigenomics & Department of Dermatology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China.
| |
Collapse
|
21
|
Wu JS, Xu CY, Mo SM, Wu XM, Du ZB, Che L, Zhang YL, Yang KL, Li TD, Ge SX, Zhang TY, Lin ZN, Lin YC. Palmitoylated COX-2 Cys555 reprogrammed mitochondrial metabolism in pyroptotic inflammatory injury in patients with post-acute COVID-19 syndrome. J Adv Res 2025:S2090-1232(25)00299-1. [PMID: 40349960 DOI: 10.1016/j.jare.2025.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 05/04/2025] [Accepted: 05/04/2025] [Indexed: 05/14/2025] Open
Abstract
INTRODUCTION The complex interplay between protein palmitoylation, mitochondrial dynamics, and inflammatory responses plays a pivotal role in respiratory diseases. One significant feature of post-acute coronavirus disease 2019 (COVID-19) syndrome (PACS) is the occurrence of a storm of inflammatory cytokines related to the NOD-like receptor protein 3 (NLRP3). However, the specific mechanisms via which palmitoylation affects mitochondrial function and its impact on the NLRP3 inflammasome under pathological respiratory conditions remain to be elucidated. OBJECTIVE This study aimed to investigate how protein palmitoylation influences inflammatory responses and mitochondrial dynamics in respiratory diseases, such as those induced by the SARS-CoV-2 spike S protein in PACS, thereby providing a therapeutic target for inflammatory lung injury. METHODS In vivo experiments were conducted using AdV5-pADM-CMV-COVID-19-S (AdV5-S) nasal drip-treated C57BL/6 mice to assess NLRP3 inflammasome activation and inflammatory response. In vitro experiments were performed using pCMV-S-transfected human lung epithelial BEAS-2B cells to analyze the effects of DHHC5-mediated palmitoylation of cyclooxygenase-2 (COX-2) at cysteine 555 (COX-2Cys555) on mitochondrial metabolism and NLRP3 inflammasome activation. RESULTS Palmitoylation of COX-2Cys555 enhanced its interaction with hexokinase 2 (HK2) to regulate mitochondrial metabolic reprogramming, leading to NLRP3 inflammasome activation and pyroptosis. Pharmacological and genetic suppression of palmitoylation diminished the mitochondrial localization of palmitoylated COX-2 and its interaction with HK2, thereby reducing mitochondrial metabolic reprogramming. Furthermore, genetic intervention targeting DHHC5 (shDhhc5) alleviated NLRP3 activation and pyroptosis, mitigating the chronic inflammatory damage associated with PACS. CONCLUSION This study highlights the regulatory role of COX-2Cys555 palmitoylation in mitochondrial metabolism and lung inflammatory injury, and suggests potential therapeutic targets to combat respiratory pathogenesis linked to palmitoylated COX-2.
Collapse
Affiliation(s)
- Jia-Shen Wu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Xiang'an Hospital of Xiamen University, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China.
| | - Chi-Yu Xu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Xiang'an Hospital of Xiamen University, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China.
| | - Su-Min Mo
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Xiang'an Hospital of Xiamen University, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China.
| | - Xin-Mou Wu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Xiang'an Hospital of Xiamen University, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China.
| | - Ze-Bang Du
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Xiang'an Hospital of Xiamen University, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China.
| | - Lin Che
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Xiang'an Hospital of Xiamen University, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China.
| | - Yi-Ling Zhang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Xiang'an Hospital of Xiamen University, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China.
| | - Kai-Li Yang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Xiang'an Hospital of Xiamen University, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China.
| | - Ting-Dong Li
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Xiang'an Hospital of Xiamen University, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China.
| | - Sheng-Xiang Ge
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Xiang'an Hospital of Xiamen University, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China.
| | - Tian-Ying Zhang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Xiang'an Hospital of Xiamen University, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China.
| | - Zhong-Ning Lin
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Xiang'an Hospital of Xiamen University, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China.
| | - Yu-Chun Lin
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Xiang'an Hospital of Xiamen University, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China.
| |
Collapse
|
22
|
Hao J, Wang M, Wu Q, Song T, Hao Y, Chang L, Hou Y, Jia Z. Deciphering the molecular mechanisms of QLQX capsules in heart failure: A multi-omics perspective. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 143:156828. [PMID: 40378592 DOI: 10.1016/j.phymed.2025.156828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 04/26/2025] [Accepted: 05/02/2025] [Indexed: 05/19/2025]
Abstract
PURPOSE This study investigates the therapeutic mechanisms of Qiliqiangxin (QLQX) capsules in treating Heart Failure with Preserved Ejection Fraction (HFpEF). The study aims to understand how QLQX impacts cardiac function and underlying molecular pathways. METHODS HFpEF was induced in a rat model through unilateral nephrectomy, DOCA pellet implantation, and a high-salt diet. Cardiac function was assessed via M-mode imaging and Doppler flow measurements, focusing on key parameters like ejection fraction and diastolic function. A network pharmacology approach identified active QLQX components and potential targets, followed by comprehensive multi-omics analyses-including transcriptomics, proteomics, and metabolomics-to uncover the molecular mechanisms modulated by QLQX. Quantitative RT-PCR was employed to measure mRNA levels of key cardiac markers, providing further insights into QLQX's impact on cardiac remodeling. RESULTS QLQX treatment significantly improved cardiac function, with notable enhancements in ejection fraction and left ventricular diastolic function. Network pharmacology revealed 530 potential targets of QLQX, with 38 overlapping HFpEF targets. Key pathways identified include cGMP-PKG, adrenergic signaling, and calcium signaling. Transcriptomic analysis showed significant gene expression changes related to inflammation, energy metabolism, and myocardial remodeling, which were reversed by QLQX. Proteomic analysis identified 401 differentially expressed proteins, enriched in pathways such as cGMP-PKG and NF-κB signaling. Metabolomic profiling highlighted the role of lipid metabolism and adrenergic signaling in HFpEF, which were normalized by QLQX. In vivo validation confirmed the involvement of the cGMP-PKG pathway, with increased serum NO and cGMP levels, improved endothelial function, and reduced pro-fibrotic markers following QLQX treatment. CONCLUSION QLQX exerts multifaceted therapeutic effects on HFpEF by modulating gene expression, protein function, and metabolic pathways, particularly through the cGMP-PKG signaling pathway. These findings support QLQX as a promising therapeutic intervention for HFpEF, offering improvements in cardiac function and reversing pathological changes at multiple molecular levels.
Collapse
Affiliation(s)
- Jiameng Hao
- China Science and Technology Development Center for Chinese Medicine, Beijing, China
| | - Mingye Wang
- College of Integrated Traditional Chinese and Western Medicine, Hebei University of Chinese Medicine, No.326, Xinshi South Road, Shijiazhuang 050091, Hebei, China
| | - Qiulan Wu
- Hebei Medical University, No.361 Zhongshan East Road, Shijiazhuang 050011, Hebei, China
| | - Tao Song
- Shijiazhuang Yiling Pharmaceutical Co., Ltd, No.238, South of Tianshan Street, Shijiazhuang 050035, Hebei, China
| | - Yuanyuan Hao
- Shijiazhuang Yiling Pharmaceutical Co., Ltd, No.238, South of Tianshan Street, Shijiazhuang 050035, Hebei, China
| | - Liping Chang
- College of Integrated Traditional Chinese and Western Medicine, Hebei University of Chinese Medicine, No.326, Xinshi South Road, Shijiazhuang 050091, Hebei, China; Hebei Medical University, No.361 Zhongshan East Road, Shijiazhuang 050011, Hebei, China; State Key Laboratory for Innovation and Transformation of Luobing Theory, Hebei, China
| | - Yunlong Hou
- College of Integrated Traditional Chinese and Western Medicine, Hebei University of Chinese Medicine, No.326, Xinshi South Road, Shijiazhuang 050091, Hebei, China; Hebei Medical University, No.361 Zhongshan East Road, Shijiazhuang 050011, Hebei, China; State Key Laboratory for Innovation and Transformation of Luobing Theory, Hebei, China.
| | - Zhenhua Jia
- Shijiazhuang Yiling Pharmaceutical Co., Ltd, No.238, South of Tianshan Street, Shijiazhuang 050035, Hebei, China; State Key Laboratory for Innovation and Transformation of Luobing Theory, Hebei, China; Hebei Yiling Hospital, Shijiazhuang, 050035, Hebei, China.
| |
Collapse
|
23
|
Dong Z, Zhang S, Zhang H, Zhao D, Pan Z, Wang D. Untargeted metabolomics for acute intra-abdominal infection diagnosis in serum and urine using UHPLC-TripleTOF MS. Front Mol Biosci 2025; 12:1534102. [PMID: 40406622 PMCID: PMC12094940 DOI: 10.3389/fmolb.2025.1534102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Accepted: 03/12/2025] [Indexed: 05/26/2025] Open
Abstract
Introduction Acute intra-abdominal infection (IAI) is a prevalent and life-threatening condition in general surgery, with significant implications for patient mortality. However, the timely identification of IAI is often hindered by the limitations of current medical laboratory sciences and imaging diagnostics. Methods To address this critical issue, we employed metabolomics to identify early biomarkers for IAI. In this study, we enrolled a cohort of 30 IAI patients and 20 healthy volunteers. Following preliminary experimental processing, all serum and urinary samples were subjected to ultrahigh performance liquid chromatography-triple time-of-flight mass spectrometry analysis. Initial metabolite profiling was conducted using total ion current chromatography and principal component analysis. Differential metabolites were subsequently identified through Student's t-test, partial least squares discriminant analysis, and support vector machine. Hierarchical clustering analysis was then applied to assess the discriminatory power of the selected metabolites. Based on receiver operating characteristic curve analysis, we identified the most promising biomarkers, which were further subjected to enrichment analysis. Additionally, we stratified patients according to the severity and etiology of IAI to explore potential differences among these subgroups. Results Our findings revealed five serum and two urinary metabolites as potential biomarkers for IAI. The serum biomarkers were associated with the Fatty Acid Biosynthesis pathway, while the urinary biomarkers were linked to the Catecholamine Biosynthesis pathway. Notably, no significant differences were observed among the three types of IAI or the seven etiologies studied. Discussion For individuals at risk of IAI, regular screening of these biomarkers could facilitate the early and convenient identification of the condition, thereby improving patient outcomes.
Collapse
Affiliation(s)
- Zhenhua Dong
- Gastric and Colorectal Surgery Department, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Shaopeng Zhang
- Gastric and Colorectal Surgery Department, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Hongwei Zhang
- Gastric and Colorectal Surgery Department, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Dingliang Zhao
- Second Urology Department, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Ziwen Pan
- Gastric and Colorectal Surgery Department, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Daguang Wang
- Gastric and Colorectal Surgery Department, The First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
24
|
Delk SC, Gurgis FW, Reddy ST. Mechanisms and applications of apolipoproteins and apolipoprotein mimetic peptides: Common pathways in cardiovascular disease and cancer. Semin Cancer Biol 2025; 113:74-84. [PMID: 40345461 DOI: 10.1016/j.semcancer.2025.05.006] [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: 02/16/2025] [Revised: 04/29/2025] [Accepted: 05/05/2025] [Indexed: 05/11/2025]
Abstract
Apolipoproteins are the defining functional component of lipoproteins and play critical roles in lipid transport and metabolism. High-density lipoprotein (HDL) and its primary functional constituent, apolipoprotein A-I, are of particular importance because of anti-inflammatory and antioxidant properties. Apolipoprotein mimetic peptides are short-chain amino acids designed to mimic the functions and alpha-helical structure of endogenous apolipoproteins and have demonstrated efficacy in ameliorating animal models of cardiovascular disease (CVD) and cancer. The mechanisms underlying the mimetics are yet to be fully elucidated, but a comprehensive review of the literature suggests that the peptides attack pathways shared in the pathophysiology of both diseases. This review also discusses the many pre-clinical studies on the mimetic peptides, highlighting possible mechanisms at work in each. Proposed mechanisms of protection against CVD and cancer include binding and removal of pro-inflammatory oxidized lipids, reduction in reactive oxygen species, and modulation of immune cell populations. Additionally, nanoparticles (NP) formulations incorporating apolipoprotein mimetic peptides or recombinant apolipoproteins have exhibited anti-atherogenic and anti-cancer activity. To date, clinical trials to assess the effect of reconstituted HDL NPs on CVD outcomes have not shown significant improvement. The large body of successful animal studies on apolipoproteins and apolipoprotein mimetic peptides presents a disconnect between pre-clinical and clinical efficacy, highlighting the need for a more complete understanding of the underlying pathways and mechanisms.
Collapse
Affiliation(s)
- Samuel C Delk
- Environmental and Molecular Toxicology Interdepartmental Degree Program, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Faheem W Gurgis
- Department of Emergency Medicine, University of Florida College of Medicine, Gainesville, Florida, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Srinivasa T Reddy
- Environmental and Molecular Toxicology Interdepartmental Degree Program, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; Division of Cardiology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA.
| |
Collapse
|
25
|
Sharma V, Sharma P, Singh TG. Therapeutic potential of COX-2 inhibitors in neuropsychiatric disorders. J Neural Transm (Vienna) 2025:10.1007/s00702-025-02932-0. [PMID: 40325255 DOI: 10.1007/s00702-025-02932-0] [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: 08/22/2024] [Accepted: 04/10/2025] [Indexed: 05/07/2025]
Abstract
Neuropsychiatric disorders such as bipolar disorder, migraine, major depressive disorder, epilepsy, attention-deficit/hyperactivity disorder, autism spectrum disorder and schizophrenia, are a huge burden on global health, impacting millions of individuals worldwide and posing significant barriers to effective treatment. Despite advancements in medication and psychotherapy, many patients continue to suffer from severe symptoms and receive little alleviation. All of these conditions are quite frequent, yet they affect people in a way that is exceedingly detrimental. The increasing evidence suggests the connection between these disorders and inflammation. Therefore, the use of anti-inflammatory agents, namely cyclooxygenase-2 (COX-2) inhibitors, offers a new approach to prevent and treat neuropsychiatric disorders. This review discusses about the COX pathway and the role of COX-2 in the neuroinflammation. Furthermore, this review highlights the COX-2 inhibitors as a promising therapeutic agent in these neuropsychiatric disorders, however, further studies are required to assess appropriate illness stage-related indication.
Collapse
Affiliation(s)
- Veerta Sharma
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India
| | - Prateek Sharma
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India
| | - Thakur Gurjeet Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India.
| |
Collapse
|
26
|
Zeng Q, Xiao W, Zhang H, Liu W, Wang X, Li Z, Han Y, Wang Z, Yan Q, Ai Q, Yang J, Li S, Ouyang W. Anti-inflammatory mechanism of Achyranthes longifolia extract and its component chikusetsusaponin IVa by modulating Nrf2/NF-κB pathways in vitro and in vivo. Fitoterapia 2025; 184:106593. [PMID: 40334821 DOI: 10.1016/j.fitote.2025.106593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 04/22/2025] [Accepted: 05/03/2025] [Indexed: 05/09/2025]
Abstract
This study aims to explore the anti-pharyngitis mechanisms of Achyranthes longifolia (Makino) Makino. extract (ALE) and the anti-inflammatory mechanisms of its major bioactive component, chikusetsusaponin IVa (CIVa). To this end, the present study established an ammonia-induced acute pharyngitis rat model to assess the therapeutic efficacy of ALE and a lipopolysaccharide (LPS)-induced RAW264.7 cells model to evaluate the anti-inflammatory and antioxidant properties of CIVa. Pharyngeal severity was evaluated using appearance index and HE staining, while ELISA was employed to quantify inflammatory cytokines TNF-α, PGE2, and IL-6. Additionally, the levels of SOD, CAT, and MDA were measured to assess antioxidant status. Western blot was conducted to analyze the expression of proteins associated with the Nrf2/NF-κB pathways. The findings indicate that ALE provides protection in the ammonia-induced acute pharyngitis rat model, as evidenced by reduced pharyngeal redness, swelling, and improved histopathological changes. CIVa, the primary constituent of ALE, demonstrates anti-inflammatory effects in LPS-induced RAW 264.7 cells by inhibiting NO production and reducing the levels of inflammatory cytokines in a dose-dependent manner. The underlying mechanism appears to involve the inhibition of the NF-κB pathway, activation of the Nrf2 pathway, modulation of oxidative stress, and reduction of pro-inflammatory cytokine release. These results position ALE and CIVa as promising alternative therapeutic agents for the management of acute pharyngitis and potentially other inflammatory disorders.
Collapse
Affiliation(s)
- Qiongli Zeng
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China; Key Laboratory of Modern Research of TCM, Education Department of Hunan Province, Changsha 410208, China
| | - Weiting Xiao
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China; Key Laboratory of Modern Research of TCM, Education Department of Hunan Province, Changsha 410208, China
| | - Heng Zhang
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China; Key Laboratory of Modern Research of TCM, Education Department of Hunan Province, Changsha 410208, China
| | - Wei Liu
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China; Key Laboratory of Modern Research of TCM, Education Department of Hunan Province, Changsha 410208, China
| | - Xionglong Wang
- Analysis of Complex Effects of Proprietary Chinese Medicine, Hunan Provincial Key Laboratory, Yongzhou 410116, China
| | - Zhen Li
- Analysis of Complex Effects of Proprietary Chinese Medicine, Hunan Provincial Key Laboratory, Yongzhou 410116, China
| | - Yue Han
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China; Key Laboratory of Modern Research of TCM, Education Department of Hunan Province, Changsha 410208, China
| | - Zhi Wang
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Qian Yan
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China; Key Laboratory of Modern Research of TCM, Education Department of Hunan Province, Changsha 410208, China
| | - Qidi Ai
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Jinwei Yang
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha 410208, China; The Second Hospital of Integrated Chinese and Western Medicine Affiliated to Hunan University of Chinese Medicine, Liuyang 410300, China
| | - Shunxiang Li
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China.
| | - Wen Ouyang
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China; Key Laboratory of Modern Research of TCM, Education Department of Hunan Province, Changsha 410208, China; The Second Hospital of Integrated Chinese and Western Medicine Affiliated to Hunan University of Chinese Medicine, Liuyang 410300, China.
| |
Collapse
|
27
|
Liotti F, Marotta M, Costanzo M, De Simone C, Zirpoli S, De Falco V, Melillo RM, Prevete N. Formyl peptide receptor 1 signaling strength orchestrates the switch from pro-inflammatory to pro-resolving responses: The way to exert its anti-angiogenic and tumor suppressor functions. Biomed Pharmacother 2025; 186:117961. [PMID: 40112515 DOI: 10.1016/j.biopha.2025.117961] [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: 12/27/2024] [Revised: 02/24/2025] [Accepted: 03/04/2025] [Indexed: 03/22/2025] Open
Abstract
The well-paced trigger of inflammation resolution following an inflammatory response is crucial for tissue homeostasis and cancer. In gastrointestinal tumors the Formyl peptide receptor 1 (FPR1) stimulates an inflammation resolution response able to restrain cancer angiogenesis and growth. A preceding inflammatory signal is necessary for the induction of the pro-resolving response. However, if FPR1-induced inflammation resolution and tumor suppressor function require an early pro-inflammatory trigger and how this is achieved remains unknown. A ROS-dependent signaling is activated in response to FPR1 activation. In colorectal carcinoma (CRC) cells, we carefully analyzed this signal showing that FPR1 activation by the fMLF peptide induces biphasic ROS production: a first wave, early, mitochondrial (mROS), followed by a second, late, NADPH oxidase (NOX1)-dependent. mROS cause SHP2 phosphatase inactivation restraining its ability to dephosphorylate and inactivate SRC. SRC, in turn, allows the activation of RAS and Rac1 GTPases. RAS activates MAPK signaling, while Rac1 supports NOX1 activation, that causes the second wave of ROS, reinforcing this signaling cycle. Importantly, for the first time, we demonstrate that mROS production precedes and is necessary for pro-inflammatory mediators' release, while NOX1-dependent ROS are only required for pro-resolving mediators' synthesis. Pharmacological and genetic approaches and functional assays show that this signaling cascade is essential for the pro-resolving and anti-angiogenic properties of FPR1 in CRC. In conclusion, we show that FPR1 elicits pro-resolving effects in CRC activating two waves of ROS production characterized by different strength and kinetics, that parallel and are necessary for pro-inflammatory or pro-resolving mediators' production.
Collapse
Affiliation(s)
- Federica Liotti
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Italy
| | - Maria Marotta
- Institute of Endotypes in Oncology, Metabolism and Immunology (IEOMI), CNR, Naples, Italy
| | - Mattia Costanzo
- Department of Translational Medical Sciences, University of Naples Federico II, Italy
| | - Chiara De Simone
- Department of Translational Medical Sciences, University of Naples Federico II, Italy
| | - Sara Zirpoli
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Italy
| | - Valentina De Falco
- Institute of Endotypes in Oncology, Metabolism and Immunology (IEOMI), CNR, Naples, Italy
| | - Rosa Marina Melillo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Italy.
| | - Nella Prevete
- Department of Translational Medical Sciences, University of Naples Federico II, Italy.
| |
Collapse
|
28
|
Chen Y, Li X, Sun X, Kou Y, Ma X, Song L, Zhang H, Xie F, Song Z, Yuan C, Huang S, Wu Y. Joint transcriptomics and metabolomics unveil the protective mechanism of tamarind seed polysaccharide against antibiotic-induced intestinal barrier damage. Int J Biol Macromol 2025; 305:140999. [PMID: 39952497 DOI: 10.1016/j.ijbiomac.2025.140999] [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: 01/13/2025] [Accepted: 02/11/2025] [Indexed: 02/17/2025]
Abstract
Intestinal barrier damage is frequently caused by antibiotic therapy, potentially leading to bacterial translocation and toxin leakage, which triggers inflammation and increases the risk of various diseases. In this study, Tamarind seed polysaccharides (TSP) with different molecular weights were administered to mice during the recovery phase from clindamycin-induced intestinal barrier damage. The results indicated that TSP restored the shortened colon length, reduced the enlarged cecum index, and decreased the elevated level of inflammatory infiltration. Biochemical testing revealed that TSP decreased the levels of intestinal permeability biomarkers and inflammatory factors that were elevated by clindamycin treatment. Transcriptomics and non-targeted metabolomics analyses respectively uncovered changes in colon gene expression and fecal metabolites. The joint analysis of these omics data identified critical pathways, including arachidonic acid metabolism, retinol metabolism, and steroid hormone biosynthesis. These findings suggest that TSP could be a promising dietary supplement for protecting the intestinal barrier and alleviating inflammation.
Collapse
Affiliation(s)
- Yinan Chen
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Xujiao Li
- Institute for Agro-food Standards and Testing Technology, Laboratory of Quality & Safety Risk Assessment for Agro-products (Shanghai), Ministry of Agriculture and Rural Affairs, Shanghai Academy of Agricultural Sciences, Shanghai 201403, China
| | - Xianbao Sun
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yuxing Kou
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Xuan Ma
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Lihua Song
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Hui Zhang
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Fan Xie
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Zibo Song
- Yunnan Maoduoli Group Food Co., Ltd., Yuxi 653100, China; Yunnan Special Favor Biotechnology Co., Ltd., Yuxi 653100, China
| | - Chunmei Yuan
- Yunnan Maoduoli Group Food Co., Ltd., Yuxi 653100, China; Yunnan Special Favor Biotechnology Co., Ltd., Yuxi 653100, China
| | - Siyan Huang
- Yunnan Maoduoli Group Food Co., Ltd., Yuxi 653100, China; Yunnan Special Favor Biotechnology Co., Ltd., Yuxi 653100, China
| | - Yan Wu
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
29
|
Iqbal U, Malik A, Sial NT, Mehmood MH, Uttra AM, Tulain UR, Erum A, Fayyaz-Ur-Rehman M, Welson NN, Mahmoud MH, Alexiou A, Papadakis M, El-Saber Bathia G. Eucalyptol attenuates indomethacin-induced gastric ulcers in rats by modulating the ICAM-1, eNOS and COX/LOX pathways: Insights from in silico, in vitro and in vivo approaches. Food Chem Toxicol 2025; 199:115319. [PMID: 39965739 DOI: 10.1016/j.fct.2025.115319] [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: 01/07/2025] [Accepted: 02/10/2025] [Indexed: 02/20/2025]
Abstract
In order to evaluate anti-inflammatory role of eucalyptol (100, 200, and 400 mg/kg orally), inflammation was induced in rats using 0.1 ml of histamine and 0.1 ml of formaldehyde. Furthermore, in vivo gastroprotective potential of eucalyptol (100, 200 and 400 mg/kg) was determined via the intraperitoneal injection of 25 mg/kg indomethacin as an ulcerative agent and omeprazole (30 mg/kg) orally as a standard. Estimation of biochemical (PGE2, ICAM-1, COX-I, COX-II, eNOS and 5-LOX) and oxidative stress (SOD, CAT, GSH, and MDA) markers were carried out in gastric tissues using ELISA. The morphological and histopathological features of the gastric tissues were studied. In vitro, eucalyptol stabilized red blood cell membranes and inhibited protein denaturation, with the maximum effect observed at a concentration of 6400 μg/mL. Eucalyptol significantly reduced rat paw edema in histamine- and formaldehyde-induced inflammation models. It increased gastric PGE2, COX-I and eNOS levels, and decreased COX-II, 5-LOX and ICAM-1. Eucalyptol reduced ulcer indices and improved histopathological changes. Eucalyptol also increased antioxidants levels with decreased MDA levels in isolated rat stomach tissues. Therefore, eucalyptol shows gastroprotective effects against histamine- and formaldehyde induced inflammation and indomethacin-induced gastric ulcers through the modulation of the COX/LOX, ICAM-1, eNOS pathways and oxidative stress biomarkers.
Collapse
Affiliation(s)
- Urooj Iqbal
- Department of Pharmacology, College of Pharmacy, University of Sargodha, Pakistan; Primary and Secondary Health Care Department, Lahore, Punjab, Pakistan.
| | - Abdul Malik
- Department of Pharmacology, College of Pharmacy, University of Sargodha, Pakistan.
| | - Nabeela Tabassum Sial
- Department of Pharmacology, College of Pharmacy, University of Sargodha, Pakistan; Institute of Pharmacy, Lahore College for Women University, Lahore, Pakistan.
| | - Malik Hassan Mehmood
- Department of Pharmaceutical Sciences, Government College University Lahore, Pakistan.
| | - Ambreen Malik Uttra
- Department of Pharmacology, College of Pharmacy, University of Sargodha, Pakistan.
| | - Ume Ruqia Tulain
- Department of Pharmaceutics, College of Pharmacy, University of Sargodha, Pakistan.
| | - Alia Erum
- Department of Pharmaceutics, College of Pharmacy, University of Sargodha, Pakistan.
| | | | - Nermeen N Welson
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Beni-Suef University, 62511, Beni Suef, Egypt.
| | - Mohamed H Mahmoud
- Department of Biochemistry, College of Science, King Saud University, Kingdom of Saudi Arabia.
| | - Athanasios Alexiou
- University Centre for Research & Development, Chandigarh University, Chandigarh-Ludhiana Highway, Mohali, Punjab, India; Department of Research & Development, Funogen, Athens, 11741, Greece.
| | - Marios Papadakis
- University Hospital Witten-Herdecke, Heusnerstrasse 40, University of Witten-Herdecke, 42283, Wuppertal, Germany.
| | - Gaber El-Saber Bathia
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, AlBeheira, Damanhour, 22511, Egypt.
| |
Collapse
|
30
|
Li J, Xuan M, Yang L, Liu Y, Lou N, Fu L, Shi Q, Xue C. Comprehensive single-cell analysis deciphered the immunoregulatory mechanism of TPPU in alleviating sepsis-related acute liver injury. J Adv Res 2025; 71:457-470. [PMID: 39956402 DOI: 10.1016/j.jare.2025.02.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 01/21/2025] [Accepted: 02/13/2025] [Indexed: 02/18/2025] Open
Abstract
INTRODUCTION Sepsis-related acute liver injury involves complex immune dysfunctions. Epoxyeicosatrienoic acids (EETs), bioactive molecules derived from arachidonic acid (AA) via cytochrome P450 (CYP450) and rapidly hydrolyzed by soluble epoxide hydrolase (sEH), possess anti-inflammatory properties. Nevertheless, the impact of the sEH inhibitor TPPU on sepsis-related acute liver injury remains uncertain. OBJECTIVES This study utilized comprehensive single-cell analysis to investigate the immunoregulatory mechanism of TPPU in alleviating sepsis-related acute liver injury. METHODS Hepatic bulk RNA sequencing and proteomics analyses were employed to investigate the mechanisms underlying sepsis-related acute liver injury induced by cecal ligation and puncture in mice. Cytometry by time-of-flight and single-cell RNA sequencing were conducted to thoroughly examine the immunoregulatory role of TPPU at single-cell resolution. RESULTS Downregulation of AA metabolism and the CYP450 pathway was observed during sepsis-related acute liver injury, and TPPU treatment reduced inflammatory cytokine production and mitigated sepsis-related hepatic inflammatory injury. Comprehensive single-cell analysis revealed that TPPU promotes the expansion of anti-inflammatory CD206+CD73+ M2-like macrophages and PDL1-CD39-CCR2+ neutrophils, reprogramming liver neutrophils to an anti-inflammatory CAMP+NGP+CD177+ phenotype. Additionally, TPPU inhibits the CCL6-CCR1 signaling mediated by M2-like macrophages and CAMP+NGP+CD177+ neutrophils, altering intercellular communication within the septic liver immune microenvironment. CONCLUSION This study demonstrated TPPU's protective efficacy against sepsis-related acute liver injury, underscoring its vital role in modulating liver macrophages and neutrophils and enhancing prospects for personalized immunomodulatory therapy.
Collapse
Affiliation(s)
- Juan Li
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Mengjuan Xuan
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Li Yang
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yingru Liu
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Na Lou
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Leiya Fu
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Qingmiao Shi
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| | - Chen Xue
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| |
Collapse
|
31
|
Xie Y, Zeng Z, Zhang J, Han Q, Song C, Jin S, Zhao M. Cuscutae Semen in depression-induced ovarian dysfunction: metabolomics with UPLC-QToF-MS in female mice. Front Mol Biosci 2025; 12:1595602. [PMID: 40370520 PMCID: PMC12074923 DOI: 10.3389/fmolb.2025.1595602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2025] [Accepted: 04/18/2025] [Indexed: 05/16/2025] Open
Abstract
The increasing prevalence of depression profoundly affects female ovarian health. Although Cuscutae Semen (CS) is acknowledged for treating reproductive disorders, its pharmacological mechanisms in depression-induced ovarian dysfunction remain insufficiently explored. This study investigated CS's effects in a chronic unpredictable mild stress (CUMS) mouse model of depression. Mice were divided into control, CUMS model, CS treatment and estradiol treatment group. Behavioral and biochemical analyses assessed depressive-like behaviors and hormone levels. Untargeted metabolomics utilizing ultra-performance liquid chromatography coupled with quadrupole time-of-flight mass spectrometry was applied to identify differential metabolites of CS in the treatment of depression-induced ovarian dysfunction. These findings were confirmed through real-time quantitative polymerase chain reaction assays. Based on the outcomes from behavioral and biochemical assays, CS effectively ameliorated the chronic unpredictable mild stress-induced reproductive ailment in mice. Ten differential metabolites were identified, highlighting the impact of CUMS and CS's ameliorative effects. Pathways linked to arachidonic acid metabolism, glycerophospholipid metabolism, linoleic acid metabolism, and steroid hormone biosynthesis were involved. Seven target genes further validated the metabolomic analysis. This study provides strong evidence of CS's therapeutic potential in alleviating depression-induced ovarian dysfunction, shedding light on its pharmacological mechanisms and supporting its use as a functional medical food.
Collapse
Affiliation(s)
- Ying Xie
- School of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan, Hubei, China
| | - Zhaoxiang Zeng
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, Hubei, China
| | - Jinrong Zhang
- School of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan, Hubei, China
| | | | - Chengwu Song
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, Hubei, China
- Hubei Shizhen Laboratory, Wuhan, Hubei, China
| | - Shuna Jin
- School of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan, Hubei, China
- Hubei Shizhen Laboratory, Wuhan, Hubei, China
| | - Min Zhao
- School of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan, Hubei, China
- Hubei Shizhen Laboratory, Wuhan, Hubei, China
| |
Collapse
|
32
|
Huang R, Yong X, Li T, Wen H, Zhou X, Liao Y, You J, Yu C, Xu P, Wang Y, Wen D, Xia T, Yang H, Chen Y, Xu L, Zhong X, Li X, Xu Z, Zhou C. 15-Lipoxygenase-2 deficiency induces foam cell formation that can be restored by salidroside through the inhibition of arachidonic acid effects. Open Life Sci 2025; 20:20251091. [PMID: 40321157 PMCID: PMC12048898 DOI: 10.1515/biol-2025-1091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 02/21/2025] [Accepted: 03/10/2025] [Indexed: 05/08/2025] Open
Abstract
15-Lipoxygenase-2 (15-Lox-2) is one of the key enzymes in arachidonic acid (AA) metabolic pathway, which belongs to the unsaturated fatty acid metabolic pathway. This pathway is involved in the foam cell transformation of macrophages during the progression of atherosclerosis (AS). The role of salidroside (SAL) in cardiovascular diseases has been extensively studied, but its impact on macrophage foam cell formation has not yet been clearly clarified. We aimed to determine the effects of 15-Lox-2 deficiency on macrophage (Ana-1 cell) foam cell formation, and those of SAL on 15-Lox-2-deficient macrophages. 15-Lox-2-deficient macrophages were generated using short hairpin RNA. Results indicated that 15-Lox-2 expression in the aorta of atherosclerotic patients is lower than that of the normal group. Additionally, 15-Lox-2 deficiency dramatically promoted macrophage uptake of oxidized low-density lipoprotein (ox-LDL) and increased the Cyclin D1 level while dramatically decreasing caspase3 expression. Furthermore, inflammation, complement, and TNF-α signaling pathways, along with IL1α, IL1β, IL18, and Cx3cl1, were activated in 15-Lox-2-deficient macrophages. These changes were alleviated by SAL through inhibiting AA effects, and the effects of AA on macrophages could be inhibited by SAL. Consistently, phospholipase A2-inhibitor arachidonyl trifluoromethyl ketone (AACOCF3) restored these changes. In summary, SAL reversed the effects of 15-Lox-2 deficiency on macrophages by inhibiting excessive AA and may be a promising therapeutic potential in treating atherosclerosis resulting from 15-Lox-2 deficiency.
Collapse
Affiliation(s)
- Rong Huang
- Institute of Materia Medica, School of Pharmacy, North Sichuan Medical College, No. 234, Fujiang Road, Nanchong, Sichuan, 637000, China
| | - Xi Yong
- Department of Vascular Surgery, Department of Nuclear Medicine, Radiotherapy Department, Department of Oncology, Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Tingting Li
- Department of Pharmacy, Second Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Huling Wen
- Department of Vascular Surgery, Department of Nuclear Medicine, Radiotherapy Department, Department of Oncology, Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Xing Zhou
- Translational Medicine Research Center, Institute of Hepatobiliary Research, School of Basic Medical Sciences, North Sichuan Medical College, Nanchong, Sichuan, China
| | - Yichen Liao
- Institute of Materia Medica, School of Pharmacy, North Sichuan Medical College, No. 234, Fujiang Road, Nanchong, Sichuan, 637000, China
| | - Jun You
- Institute of Materia Medica, School of Pharmacy, North Sichuan Medical College, No. 234, Fujiang Road, Nanchong, Sichuan, 637000, China
| | - Chunlei Yu
- Institute of Materia Medica, School of Pharmacy, North Sichuan Medical College, No. 234, Fujiang Road, Nanchong, Sichuan, 637000, China
| | - Peng Xu
- Department of Vascular Surgery, Department of Nuclear Medicine, Radiotherapy Department, Department of Oncology, Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Yuquan Wang
- Department of Vascular Surgery, Department of Nuclear Medicine, Radiotherapy Department, Department of Oncology, Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Dan Wen
- Department of Vascular Surgery, Department of Nuclear Medicine, Radiotherapy Department, Department of Oncology, Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Tianqin Xia
- Institute of Materia Medica, School of Pharmacy, North Sichuan Medical College, No. 234, Fujiang Road, Nanchong, Sichuan, 637000, China
| | - Hao Yang
- Translational Medicine Research Center, Institute of Hepatobiliary Research, School of Basic Medical Sciences, North Sichuan Medical College, Nanchong, Sichuan, China
| | - Yanqin Chen
- Translational Medicine Research Center, Institute of Hepatobiliary Research, School of Basic Medical Sciences, North Sichuan Medical College, Nanchong, Sichuan, China
| | - Lei Xu
- Translational Medicine Research Center, Institute of Hepatobiliary Research, School of Basic Medical Sciences, North Sichuan Medical College, Nanchong, Sichuan, China
| | - Xiaorong Zhong
- Translational Medicine Research Center, Institute of Hepatobiliary Research, School of Basic Medical Sciences, North Sichuan Medical College, Nanchong, Sichuan, China
| | - Xianfu Li
- Department of Vascular Surgery, Department of Nuclear Medicine, Radiotherapy Department, Department of Oncology, Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Zhengmin Xu
- Institute of Materia Medica, School of Pharmacy, North Sichuan Medical College, No. 234, Fujiang Road, Nanchong, Sichuan, 637000, China
| | - Chunyang Zhou
- Institute of Materia Medica, School of Pharmacy, North Sichuan Medical College, No. 234, Fujiang Road, Nanchong, Sichuan, 637000, China
| |
Collapse
|
33
|
Hansen CE, Konings J, Toth G, Chornyi S, Karsten M, van Het Hof B, van der Pol SMA, Beekhuis-Hoekstra SD, Kok N, Fung WK, Dijksman NS, Baron W, Witte ME, Lanekoff I, de Vries HE, Kooij G. Spatial mapping of the AA-PGE 2-EP axis in multiple sclerosis lesions. Acta Neuropathol 2025; 149:39. [PMID: 40299057 PMCID: PMC12041062 DOI: 10.1007/s00401-025-02878-3] [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: 10/18/2024] [Revised: 04/05/2025] [Accepted: 04/07/2025] [Indexed: 04/30/2025]
Abstract
Bioactive lipid mediators (LMs) derived from polyunsaturated fatty acids (PUFAs) are key molecules in both the initiation and resolution of inflammatory responses. Previous findings suggest that a dysregulated LM balance, especially within the arachidonic acid (AA) pathway, may contribute to an impaired resolution response and subsequent chronic neuroinflammation in multiple sclerosis (MS). However, to date, the local biosynthesis and signaling of LMs within the brain of people with MS (PwMS) remains unexplored. In this study, we, therefore, mapped the distribution of AA and its key downstream LM prostaglandin E2 (PGE2) in white matter MS brain tissue and of non-neurological controls (NNCs) for the first time using mass spectrometry imaging. We found that AA levels are lower in MS cases compared to NNCs and reduced in MS lesions compared to peri-lesional tissue. Furthermore, the PGE2/AA ratio, indicating the PGE2 synthesis from the AA substrate, was increased in lesion areas compared to fully myelinated regions in MS. In line with that, the expression of prostaglandin synthesizing enzymes as measured by RT-qPCR was partially increased in MS tissue compared to NNCs. In addition, the expression of prostaglandin E2 receptor 4 (EP4) decreased, while prostaglandin E2 receptor 2 (EP2) showed increased expression levels in MS lesions compared to NNCs and localized specifically to microglia. We also found that PGE2 addition to pro-inflammatory human-induced pluripotent stem cell (iPSC)-derived microglia resulted in enhanced cytokine signaling pathways, but also the upregulation of its synthase PTGES and homeostatic/resolving signaling, the latter of which might mainly occur through EP2 signaling. Collectively, our results provide detailed information about the region-specific levels of AA and PGE2 in MS lesions and we propose enhanced PGE2-EP2 signaling in inflamed microglia in MS.
Collapse
Affiliation(s)
- Cathrin E Hansen
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands
- MS Center Amsterdam, UMC Location VU Medical Center, Amsterdam, The Netherlands
| | - Julia Konings
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands
- MS Center Amsterdam, UMC Location VU Medical Center, Amsterdam, The Netherlands
| | - Gabor Toth
- Department of Chemistry, BMC, Uppsala University, 75237, Uppsala, Sweden
- Center of Excellence for the Chemical Mechanisms of Life, Uppsala University, Uppsala, Sweden
| | - Serhii Chornyi
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
| | - Manon Karsten
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
| | - Bert van Het Hof
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
| | - Susanne M A van der Pol
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
| | - Stephanie D Beekhuis-Hoekstra
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
| | - Nine Kok
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
| | - Wing Ka Fung
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
| | - Naomi S Dijksman
- Biomedical Sciences, Section Molecular Neurobiology, University of Groningen, UMCG, MS Center Noord Nederland, A. Deusinglaan 1, Groningen, The Netherlands
| | - Wia Baron
- Biomedical Sciences, Section Molecular Neurobiology, University of Groningen, UMCG, MS Center Noord Nederland, A. Deusinglaan 1, Groningen, The Netherlands
| | - Maarten E Witte
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands
- MS Center Amsterdam, UMC Location VU Medical Center, Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam UMC, Amsterdam, The Netherlands
| | - Ingela Lanekoff
- Department of Chemistry, BMC, Uppsala University, 75237, Uppsala, Sweden
- Center of Excellence for the Chemical Mechanisms of Life, Uppsala University, Uppsala, Sweden
| | - Helga E de Vries
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands
- MS Center Amsterdam, UMC Location VU Medical Center, Amsterdam, The Netherlands
| | - Gijs Kooij
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands.
- Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands.
- MS Center Amsterdam, UMC Location VU Medical Center, Amsterdam, The Netherlands.
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam UMC, Amsterdam, The Netherlands.
| |
Collapse
|
34
|
Chen J, Wang S, Ding Y, Xu D, Zheng S. Radiotherapy-induced alterations in tumor microenvironment: metabolism and immunity. Front Cell Dev Biol 2025; 13:1568634. [PMID: 40356601 PMCID: PMC12066526 DOI: 10.3389/fcell.2025.1568634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Accepted: 04/16/2025] [Indexed: 05/15/2025] Open
Abstract
Tumor metabolism plays a pivotal role in shaping immune responses within the tumor microenvironment influencing tumor progression, immune evasion, and the efficacy of cancer therapies. Radiotherapy has been shown to impact both tumor metabolism and immune modulation, often inducing immune activation through damage-associated molecular patterns and the STING pathway. In this study, we analyse the particular characteristics of the tumour metabolic microenvironment and its effect on the immune microenvironment. We also review the changes in the metabolic and immune microenvironment that are induced by radiotherapy, with a focus on metabolic sensitisation to the effects of radiotherapy. Our aim is to contribute to the development of research ideas in the field of radiotherapy metabolic-immunological studies.
Collapse
Affiliation(s)
- Jinpeng Chen
- Department of General Surgery, Zhongda Hospital Southeast University, Nanjing, Jiangsu, China
- Southeast University Medical School, Nanjing, Jiangsu, China
| | - Sheng Wang
- Department of Radiation Oncology, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing, Jiangsu, China
| | - Yue Ding
- Department of General Surgery, Zhongda Hospital Southeast University, Nanjing, Jiangsu, China
- Southeast University Medical School, Nanjing, Jiangsu, China
| | - Duo Xu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shiya Zheng
- Southeast University Medical School, Nanjing, Jiangsu, China
- Department of Oncology, Southeast University, Zhongda Hospital Southeast University, Nanjing, Jiangsu, China
| |
Collapse
|
35
|
Zhang Y, Ma R, Du X, He X, Zhang Y, Ma N, Liu H, Zhao X. Impact of bacteroides uniformis on fatty liver hemorrhagic syndrome in dawu golden phoenix laying hens: modulation of gut microbiota and arachidonic acid metabolism. Front Microbiol 2025; 16:1560887. [PMID: 40356654 PMCID: PMC12066428 DOI: 10.3389/fmicb.2025.1560887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 03/25/2025] [Indexed: 05/15/2025] Open
Abstract
This study explored the impact of Bacteroides uniformis (B. uniformis) on fatty liver hemorrhagic syndrome (FLHS) induced by a high-energy and low-protein (HELP) diet in laying hens, mainly focusing on hepatic lipid metabolism, gut microbiota, and arachidonic acid (AA) metabolism. A total of 120 Dawu Golden Phoenix laying hens (210-day-old) were randomly divided into four groups. The control group (CON) was fed a standard diet and received a daily gavage of PBS, while the other groups were fed with a HELP diet to induce FLHS and received a daily gavage of PBS (MOD), 1 × 109 CFU/ml B. uniformis (BUL), and 1 × 1011 CFU/ml B. uniformis (BUH) for 70 days. All hens were administered 1 ml daily by gavage. Each group had 6 replications with 5 hens per replication. The results showed that B. uniformis increased the egg production rate and feed conversion ratio and decreased body weight, liver index, and abdominal fat rate (p < 0.05). B. uniformis treatment reduced liver lipid accumulation by reducing the levels of Triglyceride (TG), Total cholesterol (TC), low-density lipoprotein cholesterol (LDL-C), alanine transaminases (ALT), and aspartate transaminases (AST) in serum and significantly elevated high-density lipoprotein cholesterol (HDL-C) (p < 0.05). The results indicated that B. uniformis altered the gut microbiota. Specifically, the abundance of Bacteroides was higher, and the relative abundances of Treponema, Helicobacter, and Spirochaetota were lower than those of the MOD group (p < 0.05). Moreover, targeted metabolomic analysis showed that supplementation of B. uniformis significantly elevated 6-keto-PGF1α and AA levels, along with significantly reduced levels of thromboxane B2 (TXB2), leukotriene D4 (LTD4), 8-isoprostaglandin F2α (8-iso-PGF2α), 12S-hydroxyeicosatetraenoic acid (12S-HETE), 15S-hydroxyeicosatetraenoic acid (15S-HETE), 9-S-hydroxy-octadecadienoic acid (9S-HODE), and 13-S-hydroxy-octadecadienoic acid (13S-HODE) (p < 0.05). In conclusion, the oral intake of B. uniformis can improve liver function, gut microbiota, and AA metabolism, thereby helping to ameliorate FLHS in Dawu Golden Phoenix laying hens.
Collapse
Affiliation(s)
- Yu Zhang
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, China
| | - Rongfei Ma
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, China
| | - Xicui Du
- Hebei Jinkun Animal Pharmaceutical Co. Ltd., Xinji, China
| | - Xin He
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, China
| | - Yan Zhang
- Institute of Animal Science and Veterinary Medicine, Hainan Academy of Agricultural Sciences, Haikou, China
| | - Ning Ma
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, China
| | - Hailong Liu
- Institute of Animal Science and Veterinary Medicine, Hainan Academy of Agricultural Sciences, Haikou, China
| | - Xinghua Zhao
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, China
| |
Collapse
|
36
|
Bourboula A, Mantzourani C, Chalatsa I, Machalia C, Emmanouilidou E, Kokotou MG, Kokotos G. A Lipidomic Approach to Studying the Downregulation of Free Fatty Acids by Cytosolic Phospholipase A 2 Inhibitors. Biomolecules 2025; 15:626. [PMID: 40427519 PMCID: PMC12108850 DOI: 10.3390/biom15050626] [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: 02/10/2025] [Revised: 04/24/2025] [Accepted: 04/25/2025] [Indexed: 05/29/2025] Open
Abstract
Inhibitors of cytosolic phospholipase A2 (GIVA cPLA2) have received great attention, since this enzyme is involved in a number of inflammatory diseases, including cancer and auto-immune and neurodegenerative diseases. Traditionally, the effects of GIVA cPLA2 inhibitors in cells have been studied by determining the inhibition of arachidonic acid release. However, although to a lesser extent, GIVA cPLA2 may also hydrolyze glycerophospholipids, releasing other free fatty acids (FFAs), such as linoleic acid or oleic acid. In the present work, we applied a liquid chromatography-high-resolution mass spectrometry method to study the levels of intracellular FFAs, after treating cells with selected GIVA cPLA2 inhibitors. Six inhibitors belonging to different chemical classes were studied, using SH-SY5Y neuroblastoma cells as a model. This lipidomic approach revealed that treatment with each inhibitor created a distinct intracellular FFA profile, suggesting not only inhibitory potency against GIVA cPLA2, but also other parameters affecting the outcome. Potent inhibitors were found to reduce not only arachidonic acid, but also other long-chain FAs, such as adrenic or linoleic acid, even medium-chain FAs, such as caproic or caprylic acid, suggesting that GIVA cPLA2 inhibitors may affect FA metabolic pathways in general. The downregulation of intracellular FFAs may have implications in reprogramming FA metabolism in neurodegenerative diseases and cancer.
Collapse
Affiliation(s)
- Asimina Bourboula
- Department of Chemistry, National and Kapodistrian University of Athens, 15771 Athens, Greece; (A.B.); (C.M.)
- Center of Excellence for Drug Design and Discovery, National and Kapodistrian University of Athens, 15771 Athens, Greece
| | - Christiana Mantzourani
- Department of Chemistry, National and Kapodistrian University of Athens, 15771 Athens, Greece; (A.B.); (C.M.)
- Center of Excellence for Drug Design and Discovery, National and Kapodistrian University of Athens, 15771 Athens, Greece
| | - Ioanna Chalatsa
- Laboratory of Biochemistry, Department of Chemistry, National and Kapodistrian University of Athens, 15771 Athens, Greece; (I.C.); (C.M.); (E.E.)
| | - Christina Machalia
- Laboratory of Biochemistry, Department of Chemistry, National and Kapodistrian University of Athens, 15771 Athens, Greece; (I.C.); (C.M.); (E.E.)
| | - Evangelia Emmanouilidou
- Laboratory of Biochemistry, Department of Chemistry, National and Kapodistrian University of Athens, 15771 Athens, Greece; (I.C.); (C.M.); (E.E.)
| | - Maroula G. Kokotou
- Laboratory of Chemistry, Department of Food Science and Human Nutrition, Agricultural University of Athens, Iera Odos 75, 11855 Athens, Greece
| | - George Kokotos
- Department of Chemistry, National and Kapodistrian University of Athens, 15771 Athens, Greece; (A.B.); (C.M.)
- Center of Excellence for Drug Design and Discovery, National and Kapodistrian University of Athens, 15771 Athens, Greece
| |
Collapse
|
37
|
Han J, Li J, Yao S, Wei Z, Jiang H, Xu T, Zeng J, Xu L, Han Y. GPR75: Advances, Challenges in Deorphanization, and Potential as a Novel Drug Target for Disease Treatment. Int J Mol Sci 2025; 26:4084. [PMID: 40362321 PMCID: PMC12071931 DOI: 10.3390/ijms26094084] [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: 03/03/2025] [Revised: 04/18/2025] [Accepted: 04/23/2025] [Indexed: 05/15/2025] Open
Abstract
G protein-coupled receptor 75 (GPR75), a novel member of the rhodopsin-like G protein-coupled receptor (GPCR) family, has been identified across various tissues and organs, where it contributes to biological regulation and disease progression. Recent studies suggest potential interactions between GPR75 and ligands such as 20-hydroxyeicosatetraenoic acid (20-HETE) and C-C motif chemokine ligand 5 (CCL5/RANTES); however, its definitive endogenous ligand remains unidentified, and GPR75 is currently classified as an orphan receptor by International Union of Basic and Clinical Pharmacology (IUPHAR). Research on GPR75 deorphanization has underscored its critical roles in disease models, particularly in metabolic health, glucose regulation, and stability of the nervous and cardiovascular systems. However, the signaling pathways of GPR75 across different pathological conditions require further investigation. Importantly, ongoing studies are targeting GPR75 for drug development, exploring small molecule inhibitors, antibodies, and gene silencing techniques, positioning GPR75 as a promising GPCR target for treating related diseases. This review summarizes the recent advancements in GPR75 deorphanization research, examines its functions across tissues and systems, and highlights its links to metabolic, cardiovascular, and neurological disorders, thereby providing a resource for researchers to better understand the biological functions of this receptor.
Collapse
Affiliation(s)
- Jingyi Han
- Department of Physiology, Zunyi Medical University, Zunyi 563006, China; (J.H.); (J.L.); (S.Y.); (Z.W.); (H.J.); (T.X.); (J.Z.)
| | - Jiaojiao Li
- Department of Physiology, Zunyi Medical University, Zunyi 563006, China; (J.H.); (J.L.); (S.Y.); (Z.W.); (H.J.); (T.X.); (J.Z.)
| | - Sirui Yao
- Department of Physiology, Zunyi Medical University, Zunyi 563006, China; (J.H.); (J.L.); (S.Y.); (Z.W.); (H.J.); (T.X.); (J.Z.)
| | - Zao Wei
- Department of Physiology, Zunyi Medical University, Zunyi 563006, China; (J.H.); (J.L.); (S.Y.); (Z.W.); (H.J.); (T.X.); (J.Z.)
| | - Hui Jiang
- Department of Physiology, Zunyi Medical University, Zunyi 563006, China; (J.H.); (J.L.); (S.Y.); (Z.W.); (H.J.); (T.X.); (J.Z.)
| | - Tao Xu
- Department of Physiology, Zunyi Medical University, Zunyi 563006, China; (J.H.); (J.L.); (S.Y.); (Z.W.); (H.J.); (T.X.); (J.Z.)
| | - Junwei Zeng
- Department of Physiology, Zunyi Medical University, Zunyi 563006, China; (J.H.); (J.L.); (S.Y.); (Z.W.); (H.J.); (T.X.); (J.Z.)
| | - Lin Xu
- Department of Immunology, Zunyi Medical University, Zunyi 563006, China
| | - Yong Han
- Department of Physiology, Zunyi Medical University, Zunyi 563006, China; (J.H.); (J.L.); (S.Y.); (Z.W.); (H.J.); (T.X.); (J.Z.)
| |
Collapse
|
38
|
Li Z, Guo Z, Yang Z, Yang B, Hu Y, Xie X, Zong Z, Chen Z, Zhang K, Zhao P, Li G, Yang X, Bian L. Metabolite-dependent m 6A methylation driven by mechanotransduction-metabolism-epitranscriptomics axis promotes bone development and regeneration. Cell Rep 2025; 44:115611. [PMID: 40272981 DOI: 10.1016/j.celrep.2025.115611] [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: 08/29/2024] [Revised: 03/18/2025] [Accepted: 04/03/2025] [Indexed: 04/26/2025] Open
Abstract
Intramembranous ossification, a major bone development process, begins with the condensation of precursor cells through the timely structural adaption of extracellular matrix (ECM) catering to rapid cellular morphological changes. Inspired by this, we design a highly cell-adaptable hydrogel to recapitulate an ECM-dependent mechanotransduction-metabolism-epitranscriptomics axis in mesenchymal stromal cells (MSCs). This hydrogel significantly enhances the E-cadherin-mediated cell-cell interactions of MSCs and promotes glucose uptake and tricarboxylic acid (TCA) cycle activities. We further show that elevated succinate inhibits fat mass and obesity-associated protein (FTO), a N6-methyladenosine (m6A) demethylase, thereby enhancing methyltransferase-like 3 (METTL3)-driven m6A methylation. Methylated RNA immunoprecipitation sequencing (MeRIP-seq) indicates increased m6A methylation of runt-related transcription 2 (Runx2), a key osteogenic signaling factor, promoting osteogenesis of hydrogel-delivered MSCs and bone regeneration in critical-sized bone defects. Our findings reveal the mechanism underlying the critical impact of adaptable ECM structures on tissue development and provide valuable guidance for the design of ECM-mimetic cell carriers to enhance the therapeutic outcomes of regenerative medicine.
Collapse
Affiliation(s)
- Zhuo Li
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Sha Tin, New Territories, Hong Kong 999077, China
| | - Zhengnan Guo
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou 510006, China
| | - Zhengmeng Yang
- Department of Orthopaedic and Traumatology, The Chinese University of Hong Kong, Sha Tin, New Territories, Hong Kong 999077, China
| | - Boguang Yang
- Department of Orthopaedic and Traumatology, The Chinese University of Hong Kong, Sha Tin, New Territories, Hong Kong 999077, China
| | - Yuan Hu
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou 510006, China
| | - Xian Xie
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Sha Tin, New Territories, Hong Kong 999077, China
| | - Zhixian Zong
- Department of Orthopaedic and Traumatology, The Chinese University of Hong Kong, Sha Tin, New Territories, Hong Kong 999077, China
| | - Zekun Chen
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou 510006, China
| | - Kunyu Zhang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou 510006, China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, China; Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou 510006, China
| | - Pengchao Zhao
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou 510006, China
| | - Gang Li
- Department of Orthopaedic and Traumatology, The Chinese University of Hong Kong, Sha Tin, New Territories, Hong Kong 999077, China; Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China.
| | - Xuefeng Yang
- Anhui Key Laboratory of Modern Biomanufacturing, School of Life Sciences, Anhui University, Hefei 230601, China.
| | - Liming Bian
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou 510006, China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, China; Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou 510006, China.
| |
Collapse
|
39
|
Lu Y, Chen L, Lin Y, Zhang Y, Wang Y, Yu W, Ren F, Guo H. Fatty acid metabolism: The crossroads in intestinal homeostasis and tumor. Metabolism 2025; 169:156273. [PMID: 40280478 DOI: 10.1016/j.metabol.2025.156273] [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: 03/07/2025] [Revised: 04/09/2025] [Accepted: 04/19/2025] [Indexed: 04/29/2025]
Abstract
Fatty acids (FAs) have various functions on cell regulation considering their abundant types and metabolic pathways. In addition, the relation between FA and other nutritional metabolism makes their functions more complex. As the first place for diet-derived FA metabolism, intestine is significantly influenced despite lack of clear conclusions due to the inconsistent findings. In this review, we discuss the regulation of fatty acid metabolism on the fate of intestinal stem cells in homeostasis and disorders, and also focus on the intestinal tumor development and treatment from the aspect of gut microbiota-epithelium-immune interaction. We summarize that the balances between FA oxidation and glycolysis, between oxidative phosphorylation and ketogenesis, between catabolism and anabolism, and the specific roles of individual FA types determine the diverse effects of intestinal FA metabolism in different cases. We hope this will inspire further dissection and suggest precise dietary/metabolic intervention for different demands related to intestinal health.
Collapse
Affiliation(s)
- Yao Lu
- Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Lining Chen
- Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Yingying Lin
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China
| | - Yafei Zhang
- Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Yuqi Wang
- Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Weiru Yu
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China
| | - Fazheng Ren
- Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China.
| | - Huiyuan Guo
- Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
40
|
Zhang X, Gschwind J, Erben V, Bennewitz K, Li X, Sticht C, Poschet G, Hausser I, Fleming T, Szendroedi J, Nawroth PP, Kroll J. Endogenous acrolein accumulation in akr7a3 mutants causes microvascular dysfunction due to increased arachidonic acid metabolism. Redox Biol 2025; 83:103639. [PMID: 40258306 PMCID: PMC12051060 DOI: 10.1016/j.redox.2025.103639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 04/14/2025] [Accepted: 04/14/2025] [Indexed: 04/23/2025] Open
Abstract
Acrolein (ACR) is an endogenous reactive unsaturated aldehyde that can be detoxified by the aldo-keto reductase (AKR) enzyme system. While it has been shown that accumulation of ACR is associated with several health problems, including inflammation, oxidative stress, and cardiovascular disease the study aimed to analyze whether an endogenous accumulation of ACR is causal for vascular dysfunction in an akr7a3 mutant zebrafish model. Enlargement of the hyaloid and retinal vasculature, as well as alterations in the larval pronephros and thickening of the glomerular basement membrane in the adult kidney were found upon ACR accumulation. Transcriptomic and metabolomic analyses, followed by functional validation, revealed that the up-regulation of genes controlling the arachidonic acid metabolism and activation of the leukotriene pathway are responsible for the observed microvascular changes. In conclusion, the data have identified an intrinsic function of ACR in akr7a3 mutants that activates the arachidonic acid metabolism and subsequently disrupts vascular integrity by promoting an inflammatory response. Thus, ACR is causal in the development of vascular disease.
Collapse
Affiliation(s)
- Xin Zhang
- Department of Vascular Biology, ECAS, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68167, Germany
| | - Johannes Gschwind
- Department of Vascular Biology, ECAS, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68167, Germany
| | - Vanessa Erben
- Department of Vascular Biology, ECAS, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68167, Germany
| | - Katrin Bennewitz
- Department of Vascular Biology, ECAS, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68167, Germany
| | - Xiaogang Li
- Department of Vascular Biology, ECAS, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68167, Germany
| | - Carsten Sticht
- NGS Core Facility, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68167, Germany
| | - Gernot Poschet
- Metabolomics Core Technology Platform, Centre for Organismal Studies, Heidelberg University, Heidelberg, 69120, Germany
| | - Ingrid Hausser
- Institute of Pathology IPH, EM Lab, Heidelberg University Hospital, Heidelberg, 69120, Germany
| | - Thomas Fleming
- Department of Internal Medicine I and Clinical Chemistry, Heidelberg University Hospital, Heidelberg, 69120, Germany
| | - Julia Szendroedi
- Department of Internal Medicine I and Clinical Chemistry, Heidelberg University Hospital, Heidelberg, 69120, Germany
| | - Peter Paul Nawroth
- Medical Clinic and Polyclinic II, University Hospital Dresden, Dresden, 01307, Germany
| | - Jens Kroll
- Department of Vascular Biology, ECAS, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68167, Germany.
| |
Collapse
|
41
|
Lage SL, Bricker-Holt K, Rocco JM, Rupert A, Donovan FX, Abramzon YA, Chandrasekharappa SC, McNinch C, Cook L, Amaral EP, Rosenfeld G, Dalhuisen T, Eun A, Hoh R, Fehrman E, Martin JN, Deeks SG, Henrich TJ, Peluso MJ, Sereti I. Persistent immune dysregulation and metabolic alterations following SARS-CoV-2 infection. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.04.16.25325949. [PMID: 40321289 PMCID: PMC12047922 DOI: 10.1101/2025.04.16.25325949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/08/2025]
Abstract
SARS-CoV-2 can cause a variety of post-acute sequelae including Long COVID19 (LC), a complex, multisystem disease characterized by a broad range of symptoms including fatigue, cognitive impairment, and post-exertional malaise. The pathogenesis of LC is incompletely understood. In this study, we performed comprehensive cellular and transcriptional immunometabolic profiling within a cohort that included SARS-CoV-2-naïve controls (NC, N=30) and individuals with prior COVID-19 (~4-months) who fully recovered (RC, N=38) or went on to experience Long COVID symptoms (N=58). Compared to the naïve controls, those with prior COVID-19 demonstrated profound metabolic and immune alterations at the proteomic, cellular, and epigenetic level. Specifically, there was an enrichment in immature monocytes with sustained inflammasome activation and oxidative stress, elevated arachidonic acid levels, decreased tryptophan, and variation in the frequency and phenotype of peripheral T-cells. Those with LC had increased CD8 T-cell senescence and a distinct transcriptional profile within CD4 and CD8 T-cells and monocytes by single cell RNA sequencing. Our findings support a profound and persistent immunometabolic dysfunction that follows SARS-CoV-2 which may form the pathophysiologic substrate for LC. Our findings suggest that trials of therapeutics that help restore immune and metabolic homeostasis may be warranted to prevent, reduce, or resolve LC symptoms.
Collapse
Affiliation(s)
- Silvia Lucena Lage
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda, USA
| | - Katherine Bricker-Holt
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda, USA
| | - Joseph M. Rocco
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda, USA
| | - Adam Rupert
- AIDS Monitoring Laboratory, Frederick National Laboratory for Cancer Research; Frederick, USA
| | - Frank X. Donovan
- Cancer Genetics and Comparative Genomics Branch, National Human Genome Research Institute; Bethesda, USA
| | - Yevgeniya A. Abramzon
- Cancer Genetics and Comparative Genomics Branch, National Human Genome Research Institute; Bethesda, USA
| | | | - Colton McNinch
- Bioinformatics and Computational Biosciences Branch, Office of Cyber Infrastructure and Computational Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, USA
| | - Logan Cook
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda, USA
| | - Eduardo Pinheiro Amaral
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda, USA
| | - Gabriel Rosenfeld
- Bioinformatics and Computational Biosciences Branch, Office of Cyber Infrastructure and Computational Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, USA
| | - Thomas Dalhuisen
- Department of Medicine, University of California, San Francisco; San Francisco, USA
| | - Avery Eun
- Department of Medicine, University of California, San Francisco; San Francisco, USA
| | - Rebecca Hoh
- Department of Medicine, University of California, San Francisco; San Francisco, USA
| | - Emily Fehrman
- Department of Medicine, University of California, San Francisco; San Francisco, USA
| | - Jeffrey N. Martin
- Department of Epidemiology and Biostatistics, University of California, San Francisco; San Francisco, USA
| | - Steven G. Deeks
- Department of Medicine, University of California, San Francisco; San Francisco, USA
| | - Timothy J. Henrich
- Department of Medicine, University of California, San Francisco; San Francisco, USA
| | - Michael J. Peluso
- Department of Medicine, University of California, San Francisco; San Francisco, USA
| | - Irini Sereti
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda, USA
| |
Collapse
|
42
|
Hole C, Dhamsania A, Brown C, Ryznar R. Immune Dysregulation in Depression and Anxiety: A Review of the Immune Response in Disease and Treatment. Cells 2025; 14:607. [PMID: 40277932 PMCID: PMC12025721 DOI: 10.3390/cells14080607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2025] [Revised: 04/13/2025] [Accepted: 04/15/2025] [Indexed: 04/26/2025] Open
Abstract
Rates of depression and anxiety have increased significantly in recent decades, with many patients experiencing treatment-resistant symptoms. Beyond psychiatric manifestations, these conditions are associated with heightened risks of suicide, cardiovascular disease, chronic pain, and fatigue. Emerging research suggests that neuroinflammation, immune dysregulation, and hypothalamic-pituitary-adrenal axis dysfunction contribute to their pathophysiology, often interacting bidirectionally with stress. While current first-line treatments primarily target neurotransmitter imbalances, many patients do not achieve symptom resolution, highlighting the need for novel approaches. This review explores the role of immune dysfunction, cytokine activity, and neurotransmitter interactions in depression and anxiety. Additionally, we examine how existing pharmacological and non-pharmacological interventions influence inflammation and immune responses. Understanding these mechanisms may pave the way for more integrative treatment strategies that combine immune modulation with traditional psychiatric therapies.
Collapse
Affiliation(s)
- Christopher Hole
- College of Osteopathic Medicine, Rocky Vista University, Englewood, CO 80112, USA; (C.H.); (A.D.); (R.R.)
| | - Akash Dhamsania
- College of Osteopathic Medicine, Rocky Vista University, Englewood, CO 80112, USA; (C.H.); (A.D.); (R.R.)
| | - Cassandra Brown
- College of Osteopathic Medicine, Rocky Vista University, Englewood, CO 80112, USA; (C.H.); (A.D.); (R.R.)
| | - Rebecca Ryznar
- College of Osteopathic Medicine, Rocky Vista University, Englewood, CO 80112, USA; (C.H.); (A.D.); (R.R.)
- Department of Biomedical Sciences, Rocky Vista University, Englewood, CO 80112, USA
| |
Collapse
|
43
|
Yuan C, Tsang A, Berumen M, Rodriguez A, Yun F, Mesic A, Olivares A, Dubon L, Nguyen A, Pavana L, Mercado M, Gorostiza G, Morisseau C, Hammock BD, Kandasamy R, Pecic S. Structure-activity relationship studies and pharmacological evaluation of 4-phenylthiazoles as dual soluble epoxide hydrolase/fatty acid amide hydrolase inhibitors. Bioorg Med Chem 2025; 121:118112. [PMID: 39983408 PMCID: PMC11910963 DOI: 10.1016/j.bmc.2025.118112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 02/06/2025] [Accepted: 02/11/2025] [Indexed: 02/23/2025]
Abstract
Forty-two 4-phenylthiazole analogs, organized in two libraries 4a-u and 6a-u, were prepared and biologically evaluated in human fatty acid amide hydrolase (FAAH), and human, rat and mouse soluble epoxide hydrolase (sEH) inhibition assays. This structure-activity relationship (SAR) study explores the impact of electronic and steric changes on the molecule's potency and binding affinity to better understand the structural features important for dual sEH/FAAH inhibition which will guide the development of novel treatments for pain and inflammation. Our SAR revealed that electron-donating groups on the aromatic ring of the 4-phenylthiazole moiety are particularly well tolerated by both enzymes when placed at the ortho, meta and para positions; however, the overall 3D shape of the molecule is very important for the potent FAAH inhibition, suggesting more restricted size of the FAAH binding pocket compared to sEH binding pocket. Two selected dual inhibitors, 4p and 4s, were tested in the rat liver microsomes stability assays and evaluated in vivo in the formalin test. Systemic administration of 4p and 4s via intraperitoneal injection decreased nociceptive behavior (i.e., licking of the injected paw) in male rats, and this effect was dose-dependent for both compounds. Two doses, 1 and 3 mg/kg of 4p, decreased nociceptive behavior to a similar extent to that of 30 mg/kg ketoprofen, a traditional nonsteroidal anti-inflammatory drug. However, only 3 mg/kg of 4s decreased nociceptive behavior compared to vehicle-treated animals, and this effect was comparable to ketoprofen-treated animals. Taken together, these findings reveal the antinociceptive potential of 4-phenylthiazole-based dual FAAH and sEH inhibitors and suggest pharmacodynamic differences within this class of inhibitors despite similar potencies in vitro.
Collapse
Affiliation(s)
- Cassandra Yuan
- Department of Chemistry & Biochemistry, California State University, Fullerton, 800 N. State College, Fullerton, CA 92834, United States
| | - Amanda Tsang
- Department of Chemistry & Biochemistry, California State University, Fullerton, 800 N. State College, Fullerton, CA 92834, United States
| | - Manuel Berumen
- Department of Chemistry & Biochemistry, California State University, Fullerton, 800 N. State College, Fullerton, CA 92834, United States
| | - Adriana Rodriguez
- Department of Chemistry & Biochemistry, California State University, Fullerton, 800 N. State College, Fullerton, CA 92834, United States
| | - Faye Yun
- Department of Chemistry & Biochemistry, California State University, Fullerton, 800 N. State College, Fullerton, CA 92834, United States
| | - Anesa Mesic
- Department of Chemistry & Biochemistry, California State University, Fullerton, 800 N. State College, Fullerton, CA 92834, United States
| | - Annie Olivares
- Department of Chemistry & Biochemistry, California State University, Fullerton, 800 N. State College, Fullerton, CA 92834, United States
| | - Lissette Dubon
- Department of Chemistry & Biochemistry, California State University, Fullerton, 800 N. State College, Fullerton, CA 92834, United States
| | - Allen Nguyen
- Department of Chemistry & Biochemistry, California State University, Fullerton, 800 N. State College, Fullerton, CA 92834, United States
| | - Lucy Pavana
- Department of Psychology, California State University, East Bay, 25800 Carlos Bee Blvd. Science S229, Hayward, CA 94542, United States
| | - Madison Mercado
- Department of Psychology, California State University, East Bay, 25800 Carlos Bee Blvd. Science S229, Hayward, CA 94542, United States
| | - Gabrielle Gorostiza
- Department of Psychology, California State University, East Bay, 25800 Carlos Bee Blvd. Science S229, Hayward, CA 94542, United States
| | - Christophe Morisseau
- Department of Entomology and Nematology, and UCD Comprehensive Cancer Center, University of California Davis, Davis, CA 95616, United States
| | - Bruce D Hammock
- Department of Entomology and Nematology, and UCD Comprehensive Cancer Center, University of California Davis, Davis, CA 95616, United States
| | - Ram Kandasamy
- Department of Psychology, California State University, East Bay, 25800 Carlos Bee Blvd. Science S229, Hayward, CA 94542, United States.
| | - Stevan Pecic
- Department of Chemistry & Biochemistry, California State University, Fullerton, 800 N. State College, Fullerton, CA 92834, United States.
| |
Collapse
|
44
|
Varghese M, Thekkelnaycke R, Soni T, Zhang J, Maddipati K, Singer K. Sex differences in the lipid profiles of visceral adipose tissue with obesity and gonadectomy. J Lipid Res 2025; 66:100803. [PMID: 40245983 DOI: 10.1016/j.jlr.2025.100803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 03/05/2025] [Accepted: 04/10/2025] [Indexed: 04/19/2025] Open
Abstract
In obesity, adipose tissue (AT) expansion is accompanied by chronic inflammation. Altered lipid composition in the visceral or gonadal white AT (GWAT) directly drive AT macrophage accumulation and activation to a proinflammatory phenotype. Sex steroid hormones modulate visceral versus subcutaneous lipid accumulation that correlates with metabolic syndrome, especially in men and postmenopausal women who are more prone to abdominal obesity. Prior studies demonstrated sex differences in GWAT lipid species in HFD-fed mice, but the role of sex hormones is still unclear. We hypothesized that sex hormone alterations with gonadectomy (GX) would further impact lipid composition in the obese GWAT. Untargeted lipidomics of obese GWAT identified sex differences in phospholipids, sphingolipids, sterols, fatty acyls, saccharolipids and prenol lipids. Males had significantly more precursor fatty acids (palmitic, oleic, linoleic, and arachidonic acid) than females and GX mice. Targeted lipidomics for fatty acids and oxylipins in the HFD-fed male and female GWAT stromal vascular fraction identified higher omega-6 to omega-3 free fatty acid profile in males and differences in PUFAs-derived prostaglandins, thromboxanes, and leukotrienes. Both obese male and female GWAT stromal vascular fraction showed increased levels of arachidonic acid-derived oxylipins compared to their lean counterparts. Bulk RNA-seq of sorted GWAT AT macrophages highlighted sex and diet differences in PUFA and oxylipin metabolism genes. These findings of sexual dimorphism in both stored lipid species and PUFA-derived mediators with diet and GX emphasize sex differences in lipid metabolism pathways that drive inflammation responses and metabolic disease risk in obesity.
Collapse
Affiliation(s)
- Mita Varghese
- Department of Pediatrics, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Rajendiran Thekkelnaycke
- Michigan Regional Comprehensive Metabolomics Resource Core, University of Michigan, Ann Arbor, MI, USA
| | - Tanu Soni
- Michigan Regional Comprehensive Metabolomics Resource Core, University of Michigan, Ann Arbor, MI, USA
| | - Jiayu Zhang
- Michigan Regional Comprehensive Metabolomics Resource Core, University of Michigan, Ann Arbor, MI, USA
| | | | - Kanakadurga Singer
- Department of Pediatrics, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
45
|
Karpurapu M, Yan J, Chung S, Pannu SR, Parinandi N, Berdyshev E, Zhang L, Christman JW. Specialized Pro-Resolving Mediator loaded Extracellular Vesicles Mitigate Pulmonary Inflammation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.09.648009. [PMID: 40291690 PMCID: PMC12027339 DOI: 10.1101/2025.04.09.648009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Specialized pro-resolving mediators (SPMs), including lipoxins derived from arachidonic acid and resolvins, protectins, and maresins derived from docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA), orchestrate the active resolution of inflammation. These SPMs are biosynthesized through the coordinated interaction of various cells in a process known as transcellular biosynthesis, involving the sequential action of cyclooxygenase-2 (COX-2), 5-lipoxygenase (5-LOX), 12-lipoxygenase (12-LOX), and/or 15-lipoxygenase (15-LOX) enzymes. Additionally, Aspirin-triggered Resolvins are produced by acetylated COX-2, along with various lipoxygenases. Although SPMs regulate various cellular processes to actively resolve inflammation, their in vivo levels are typically low. To address this limitation, we engineered a multigene expression vector that co-expresses COX-2, 5-LOX, and 15-LOX, potentiating the synthesis of various SPMs. HEK293T cells transfected with this vector and cultured with fatty acid-free BSA-complexed DHA, EPA, and aspirin, successfully mimicked both transcellular and aspirin-triggered biosynthesis of Resolvins. These Resolvins are packaged into extracellular vesicles, which significantly inhibited neutrophil adhesion to endothelial cells, preserved endothelial monolayer barrier integrity, suppressed NF-κB reporter activity, and enhanced macrophage efferocytosis in vitro . Notably, post-injury administration of Resolvin-loaded EVs mitigated pulmonary inflammation in LPS-treated mice without causing systemic or pulmonary toxicity. In summary, we report a novel cell-based platform for generating Resolvin-loaded EVs that mitigate pulmonary inflammation in mouse models, underscoring their potential for treating other acute inflammatory diseases.
Collapse
|
46
|
Rouskas K, Bocher O, Simistiras A, Emmanouil C, Mantas P, Skoulakis A, Park YC, Dimopoulos A, Glentis S, Kastenmüller G, Zeggini E, Dimas AS. Periodic dietary restriction of animal products induces metabolic reprogramming in humans with effects on cardiometabolic health. NPJ METABOLIC HEALTH AND DISEASE 2025; 3:14. [PMID: 40225784 PMCID: PMC11981922 DOI: 10.1038/s44324-025-00057-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 03/02/2025] [Indexed: 04/15/2025]
Abstract
Dietary interventions constitute powerful approaches for disease prevention and treatment. However, the molecular mechanisms through which diet affects health remain underexplored in humans. Here, we compare plasma metabolomic and proteomic profiles between dietary states for a unique group of individuals who alternate between omnivory and restriction of animal products for religious reasons. We find that short-term restriction drives reductions in levels of lipid classes and of branched-chain amino acids, not detected in a control group of individuals, and results in metabolic profiles associated with decreased risk for all-cause mortality. We show that 23% of proteins whose levels are affected by dietary restriction are druggable targets and reveal that pro-longevity hormone FGF21 and seven additional proteins (FOLR2, SUMF2, HAVCR1, PLA2G1B, OXT, SPP1, HPGDS) display the greatest magnitude of change. Through Mendelian randomization we demonstrate potentially causal effects of FGF21 and HAVCR1 on risk for type 2 diabetes, of HPGDS on BMI, and of OXT on risk for lacunar stroke. Collectively, we find that restriction-associated reprogramming improves metabolic health and emphasise high-value targets for pharmacological intervention.
Collapse
Affiliation(s)
- Konstantinos Rouskas
- Institute for Bioinnovation, Biomedical Sciences Research Center ‘Alexander Fleming’, Fleming 34, 16672 Vari, Greece
- Institute of Applied Biosciences, Centre for Research & Technology Hellas, Thessaloniki, Greece
| | - Ozvan Bocher
- Institute of Translational Genomics, Helmholtz Zentrum München – German Research Center for Environmental Health, Neuherberg, Germany
| | - Alexandros Simistiras
- Institute for Bioinnovation, Biomedical Sciences Research Center ‘Alexander Fleming’, Fleming 34, 16672 Vari, Greece
| | - Christina Emmanouil
- Institute for Bioinnovation, Biomedical Sciences Research Center ‘Alexander Fleming’, Fleming 34, 16672 Vari, Greece
| | - Panagiotis Mantas
- Institute for Bioinnovation, Biomedical Sciences Research Center ‘Alexander Fleming’, Fleming 34, 16672 Vari, Greece
| | - Anargyros Skoulakis
- Institute for Bioinnovation, Biomedical Sciences Research Center ‘Alexander Fleming’, Fleming 34, 16672 Vari, Greece
| | - Young-Chan Park
- Institute of Translational Genomics, Helmholtz Zentrum München – German Research Center for Environmental Health, Neuherberg, Germany
| | - Alexandros Dimopoulos
- Institute for Bioinnovation, Biomedical Sciences Research Center ‘Alexander Fleming’, Fleming 34, 16672 Vari, Greece
| | - Stavros Glentis
- Institute for Bioinnovation, Biomedical Sciences Research Center ‘Alexander Fleming’, Fleming 34, 16672 Vari, Greece
| | - Gabi Kastenmüller
- Institute of Computational Biology, Helmholtz Zentrum München – German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Eleftheria Zeggini
- Institute of Translational Genomics, Helmholtz Zentrum München – German Research Center for Environmental Health, Neuherberg, Germany
- Technical University of Munich (TUM) and Klinikum Rechts der Isar, TUM School of Medicine and Health, Munich, Germany
| | - Antigone S. Dimas
- Institute for Bioinnovation, Biomedical Sciences Research Center ‘Alexander Fleming’, Fleming 34, 16672 Vari, Greece
- Institute of Translational Genomics, Helmholtz Zentrum München – German Research Center for Environmental Health, Neuherberg, Germany
| |
Collapse
|
47
|
Liu C, Qiao H, Li H, Hu X, Yan M, Fu Z, Zhang H, Wang Y, Du N. Exploring the role of LOX family in glioma progression and immune modulation. Front Immunol 2025; 16:1512186. [PMID: 40270974 PMCID: PMC12014642 DOI: 10.3389/fimmu.2025.1512186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 03/11/2025] [Indexed: 04/25/2025] Open
Abstract
Background Glioma is a major cause of mortality among central nervous system tumors, with a generally poor prognosis. The lysyl oxidase (LOX) family, a group of copper-dependent amine oxidases, has been implicated in the progression of various cancers, but its specific role in glioma and its relationship with immune infiltration remains insufficiently explored. This study aims to investigate the LOX family's expression, prognostic significance, and immune infiltration dynamics in glioma to identify potential therapeutic targets. Methods A comprehensive analysis was conducted using public databases to assess gene expression, mutation frequency, and immune infiltration patterns related to the LOX family in glioma. The results were validated through survival analysis and immunohistochemistry. Functional assays, including EdU, Transwell, and flow cytometry, were used to evaluate glioma cell proliferation, migration, invasion, and apoptosis. Co-culture experiments with immune cells, ELISA, and a glioma transplantation model were employed to study the immune-modulatory effects of the LOX family. Gene and protein expression levels were further analyzed using qRT-PCR and Western blotting. Results The LOX family was significantly upregulated in low-grade gliomas and strongly associated with poor clinical outcomes. Although mutation frequencies were low, the LOX family contributed to glioma progression through pathways involving metastasis, hypoxia response, angiogenesis, and immune cell infiltration. LOX expression correlated with increased infiltration of macrophages and eosinophils and decreased presence of Treg and CD8+ T cells. Knockdown of LOX genes impaired glioma cell functions, induced apoptosis, and altered immune cell behavior by reducing M2 macrophage polarization and enhancing CD8+ T cell activity. Conclusions The LOX family is overexpressed in glioma and is associated with poor prognosis and altered immune infiltration patterns. These findings highlight the LOX family as a promising prognostic marker and therapeutic target, particularly for enhancing the effectiveness of immunotherapy in glioma treatment.
Collapse
Affiliation(s)
- Chen Liu
- Medical School of Chinese People’s Liberation Army (PLA), Beijing, China
- Department of Oncology, the Fifth Medical Center, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- Department of Radiotherapy, Air Force Medical Center, The Fourth Military Medical University, People’s Liberation Army (PLA), Beijing, China
| | - Huilian Qiao
- Department of Pathology, Air Force Medical Center, The Fourth Military Medical University, People’s Liberation Army (PLA), Beijing, China
| | - Hongqi Li
- Department of Radiotherapy, Air Force Medical Center, The Fourth Military Medical University, People’s Liberation Army (PLA), Beijing, China
| | - Xiaolong Hu
- Department of Radiation Oncology, Beijing Geriatric Hospital, Beijing, China
| | - Maohui Yan
- Department of Radiotherapy, Air Force Medical Center, The Fourth Military Medical University, People’s Liberation Army (PLA), Beijing, China
| | - Zhiguang Fu
- Department of Radiotherapy, Air Force Medical Center, The Fourth Military Medical University, People’s Liberation Army (PLA), Beijing, China
| | - Hengheng Zhang
- Department of Radiotherapy, Air Force Medical Center, The Fourth Military Medical University, People’s Liberation Army (PLA), Beijing, China
| | - Yingjie Wang
- Department of Radiotherapy, Air Force Medical Center, The Fourth Military Medical University, People’s Liberation Army (PLA), Beijing, China
| | - Nan Du
- Medical School of Chinese People’s Liberation Army (PLA), Beijing, China
- Department of Radiotherapy, Air Force Medical Center, The Fourth Military Medical University, People’s Liberation Army (PLA), Beijing, China
| |
Collapse
|
48
|
Takasugi M, Arai H, Sugimoto K, Tsushima T, Hosomi R, Fukunaga K, Takahashi K. Lysophosphatidylcholine, rich in n-3 polyunsaturated fatty acids, inhibits leukotriene production by mouse mast cells. Lipids 2025. [PMID: 40170573 DOI: 10.1002/lipd.12443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 03/18/2025] [Accepted: 03/18/2025] [Indexed: 04/03/2025]
Abstract
In fishery industries, large amounts of byproducts are discarded during processing; therefore, their use as a source of n-3 polyunsaturated fatty acids (PUFAs) is beneficial. In this study, we evaluated the potential anti-allergic effect of lysophosphatidylcholine (Lyso-PtdCho) derived from squid skin phosphatidylcholine (PtdCho) by measuring the production of leukotriene (LT) B4 and C4, which are chemical mediators produced by mast cells in immediate allergic reactions. Squid Lyso-PtdCho, with docosahexaenoic acid exclusively esterified at the sn-2 position, significantly inhibited LT production, whereas the original PtdCho obtained from squid skin showed no inhibitory activity. Lyso-PtdCho prepared from starfish viscera PtdCho, which exclusively binds eicosapentaenoic acid at the sn-2 position, also inhibited LT production, although the effect was moderate compared with that of the squid Lyso-PtdCho. It has been suggested that Lyso-PtdCho containing n-3 PUFA may alleviate allergic symptoms and contribute to the effective utilization of fishery wastes and processing byproducts.
Collapse
Affiliation(s)
- Mikako Takasugi
- Faculty of Life Science, Kyushu Sangyo University, Fukuoka, Japan
| | - Hirofumi Arai
- School of Regional Innovation and Social Design Engineering, Kitami Institute of Technology, Hokkaido, Japan
| | - Koki Sugimoto
- Faculty of Food and Nutritional Sciences, Toyo University, Saitama, Japan
| | - Tadahiro Tsushima
- Oleo Fine Chemical Division, Bizen Chemical Co., Ltd, Okayama, Japan
| | - Ryota Hosomi
- Department of Life Science and Biotechnology, Kansai University, Osaka, Japan
| | - Kenji Fukunaga
- Department of Life Science and Biotechnology, Kansai University, Osaka, Japan
| | | |
Collapse
|
49
|
Alammari AH, Isse FA, O'Croinin C, Davies NM, El-Kadi AOS. Effect of Cannabistilbene I in Attenuating Angiotensin II-Induced Cardiac Hypertrophy: Insights into Cytochrome P450s and Arachidonic Acid Metabolites Modulation. Cannabis Cannabinoid Res 2025; 10:277-288. [PMID: 39324890 DOI: 10.1089/can.2024.0148] [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: 09/27/2024] Open
Abstract
Introduction: This research investigated the impact of Cannabistilbene I on Angiotensin II (Ang II)-induced cardiac hypertrophy and its potential role in cytochrome P450 (CYP) enzymes and arachidonic acid (AA) metabolic pathways. Cardiac hypertrophy, a response to increased stress on the heart, can lead to severe cardiovascular diseases if not managed effectively. CYP enzymes and AA metabolites play critical roles in cardiac function and hypertrophy, making them important targets for therapeutic intervention. Methods: Adult human ventricular cardiomyocyte cell line (AC16) was cultured and treated with Cannabistilbene I in the presence and absence of Ang II. The effects on mRNA expression related to cardiac hypertrophic markers and CYP were analyzed using real-time polymerase chain reaction, while CYP protein levels were measured by Western blot analysis. AA metabolites were quantified using liquid chromatography-tandem mass spectrometry (LC-MS/MS). Results: Results showed that Ang II triggered hypertrophy, as evidenced by the increase in hypertrophic marker expression, and enlarged the cell surface area, effects that were alleviated by Cannabistilbene I. Gene expression analysis indicated that Cannabistilbene I upregulated CYP1A1, leading to increased enzymatic activity, as evidenced by 7-ethoxyresorufin-O-deethylase assay. Furthermore, LC-MS/MS analysis of AA metabolites revealed that Ang II elevated midchain (R/S)-hydroxyeicosatetraenoic acid (HETE) concentrations, which were reduced by Cannabistilbene I. Notably, Cannabistilbene I selectively increased 19(S)-HETE concentration and reversed the Ang II-induced decline in 19(S)-HETE, suggesting a unique protective role. Conclusion: This study provides new insights into the potential of Cannabistilbene I in modulating AA metabolites and reducing Ang II-induced cardiac hypertrophy, revealing a new candidate as a therapeutic agent for cardiac hypertrophy.
Collapse
Affiliation(s)
- Ahmad H Alammari
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Canada
| | - Fadumo Ahmed Isse
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Canada
| | - Conor O'Croinin
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Canada
| | - Neal M Davies
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Canada
| | - Ayman O S El-Kadi
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Canada
| |
Collapse
|
50
|
Tredicine M, Mucci M, Recchiuti A, Mattoscio D. Immunoregulatory mechanisms of the arachidonic acid pathway in cancer. FEBS Lett 2025; 599:927-951. [PMID: 39973474 PMCID: PMC11995684 DOI: 10.1002/1873-3468.70013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 01/10/2025] [Accepted: 01/27/2025] [Indexed: 02/21/2025]
Abstract
The arachidonic acid (AA) pathway promotes tumor progression by modulating the complex interactions between cancer and immune cells within the microenvironment. In this Review, we summarize the knowledge acquired thus far concerning the intricate mechanisms through which eicosanoids either promote or suppress the antitumor immune response. In addition, we will discuss the impact of eicosanoids on immune cells and how they affect responsiveness to immunotherapy, as well as potential strategies for manipulating the AA pathway to improve anticancer immunotherapy. Understanding the molecular pathways and mechanisms underlying the role played by AA and its metabolites in tumor progression may contribute to the development of more effective anticancer immunotherapies.
Collapse
Affiliation(s)
- Maria Tredicine
- Department of Medical, Oral and Biotechnological SciencesUniversity of Chieti‐PescaraItaly
- Center for Advanced Studies and TechnologyUniversity of Chieti‐PescaraItaly
| | - Matteo Mucci
- Department of Medical, Oral and Biotechnological SciencesUniversity of Chieti‐PescaraItaly
- Center for Advanced Studies and TechnologyUniversity of Chieti‐PescaraItaly
| | - Antonio Recchiuti
- Department of Medical, Oral and Biotechnological SciencesUniversity of Chieti‐PescaraItaly
- Center for Advanced Studies and TechnologyUniversity of Chieti‐PescaraItaly
| | - Domenico Mattoscio
- Department of Medical, Oral and Biotechnological SciencesUniversity of Chieti‐PescaraItaly
- Center for Advanced Studies and TechnologyUniversity of Chieti‐PescaraItaly
| |
Collapse
|