1
|
Lu J, Yao T, Fu S, Fan S, Ye L. Time- and concentration-dependent metabolic responses reveal adaptation failure in cadmium-exposed Haliotis diversicolor. JOURNAL OF HAZARDOUS MATERIALS 2025; 489:137578. [PMID: 39952134 DOI: 10.1016/j.jhazmat.2025.137578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 01/13/2025] [Accepted: 02/09/2025] [Indexed: 02/17/2025]
Abstract
This study elucidated the time- and concentration-dependent effects of Cd exposure on adult Haliotis diversicolor through an integrated analysis of survival, physiological indices, and metabolomic profiles. Abalones were subjected to varying Cd concentrations for one and two months. While one-month exposure yielded no significant mortality, prolonged exposure to high Cd concentrations for two months substantially elevated mortality risk (hazard ratio = 3.359). Integrated biomarker response values exhibited progressive elevation with both concentration and exposure duration, indicating cumulative perturbation of antioxidant defense and metabolic enzyme systems. Metabolomic analysis revealed distinct temporal and concentration-dependent responses. After one month of exposure, medium concentrations predominantly affected energy and amino acid metabolism with a shift toward anaerobic pathways, while high concentrations activated additional detoxification and stress response mechanisms; after two months of exposure, divergent metabolic strategies emerged, where low concentrations induced lipid remodeling, medium concentrations maintained basic metabolic adjustments, while high concentrations precipitated comprehensive cellular dysfunction. Notably, the observed metabolic focusing at two months represented a forced trade-off under allostatic overload rather than successful adaptation, particularly in high-concentration groups. Combined analysis demonstrated fundamental reorganization of cellular metabolism under prolonged exposure. These findings emphasize the importance of long-term, multi-parameter assessments in evaluating marine ecotoxicological impacts of metal pollution.
Collapse
Affiliation(s)
- Jie Lu
- Key Laboratory of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou 510300, China; Sanya Tropical Fisheries Research Institute, Sanya 572426, China
| | - Tuo Yao
- Key Laboratory of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou 510300, China; Sanya Tropical Fisheries Research Institute, Sanya 572426, China; Tropical Fisheries Research and Development Center, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Sanya 572018, China; Shenzhen Base of South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Shenzhen 518121, China
| | - Shengli Fu
- Key Laboratory of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou 510300, China
| | - Sigang Fan
- Key Laboratory of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou 510300, China
| | - Lingtong Ye
- Key Laboratory of Aquatic Product Processing, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou 510300, China.
| |
Collapse
|
2
|
He Q, Yang Q, Wu L, He Y, Zeng N, Wang Z. Neurotoxic effects of per- and polyfluoroalkyl substances (PFAS) mixture exposure in mice: Accumulations in brain and associated changes of behaviors, metabolome, and transcriptome. JOURNAL OF HAZARDOUS MATERIALS 2025; 489:137699. [PMID: 39987740 DOI: 10.1016/j.jhazmat.2025.137699] [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: 10/19/2024] [Revised: 01/22/2025] [Accepted: 02/19/2025] [Indexed: 02/25/2025]
Abstract
Humans are exposed to complex per- and polyfluoroalkyl substances (PFAS) mixtures, yet their neurotoxicity and mechanisms remains unclear. This study exposed male mice to 17 PFAS mixtures at low levels (0.2-20 µg/L) for 49 days via drinking water. Perfluoropentanoic acid (PFPeA), perfluoroheptanoic acid (PFHpA), 6:2 fluorotelomer sulfonic acid (6:2 FTS), and perfluorooctane Sulfonate (PFOS) accumulated in brain tissues, with brain/plasma ratios of 2.03-5.87, 2.94-12.88, 1.90-3.19, and 0.62-0.93, respectively. Electroencephalogram (EEG) results showed significant alterations, including a reduction in beta spectral edge (21.47-13.85 Hz) and an increase in gamma spectral edge (57.64-79.07 Hz). Histopathological analysis revealed necrosis in the hippocampus, contributing to the observed anxiety-like behaviors and memory impairments in exposed mice. Plasma metabolomics highlighted disrupted osmoprotectants, impaired glutamatergic synapse function, and tryptophan metabolism. Brain metabolomics demonstrated suppression of purine metabolism and activation of arachidonic acid metabolism, suggesting involvement in neurotoxic effects. Transcriptomic profiling further identified dysregulation in neuroactive ligand-receptor interactions, cholinergic and GABAergic synapses, and calcium signaling pathways, with oxytocin signaling highlighted as a critical mechanism. This study, for the first time, links PFAS mixture to neurotoxicity via neurotransmitter-related pathways, underscoring the need for public health policies and preventive strategies to mitigate PFAS exposure risks.
Collapse
Affiliation(s)
- Qiurong He
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, PR China
| | - Qingkun Yang
- West China School of Public Health, Sichuan University, Chengdu 610041, PR China
| | - Lin Wu
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, PR China
| | - Yuhang He
- West China School of Public Health, Sichuan University, Chengdu 610041, PR China
| | - Ni Zeng
- West China School of Medicine, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Zhenglu Wang
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, PR China.
| |
Collapse
|
3
|
Wang L, Jiang B, Ji X, Tu J, Lu F, Yang C, Zhong X, Wang L, Cai X, Yi F, He Z, Xie L, Zhou J. Sex shapes phenotype-linked metabolic signatures of stress exposure in the mouse hypothalamus and pituitary. Neurobiol Dis 2025; 209:106898. [PMID: 40185250 DOI: 10.1016/j.nbd.2025.106898] [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: 04/02/2025] [Accepted: 04/02/2025] [Indexed: 04/07/2025] Open
Abstract
In chronic stress-induced anxiodepression, sex differences in the dysfunction of the hypothalamic-pituitary-adrenal (HPA) axis are well-documented, yet the underlying molecular mechanisms remain largely unexplored. This study investigated sex-specific metabolic signatures associated with stress exposure in the hypothalamus and pituitary, given the potential significance of brain metabolism in sex-related mechanisms underlying anxiodepression. Utilizing a chronic restraint stress (CRS) model, we conducted a comparative analysis of the metabolic profiles in female and male mice to identify distinct phenotypic expressions related to sex differences. Our findings revealed that metabolite alterations in the pituitary were more pronounced than those in the hypothalamus, indicating significant sex-based variations. These differences facilitated phenotypic differentiation and underscored the relevance of sex-specific metabolic changes and their functional associations to behavioral phenotypes. Moreover, diverging and converging pathways were identified to elucidate the molecular and physiological bases of stress susceptibility in both sexes. Key metabolic and immune-related pathways in the hypothalamus and pituitary, such as histidine, tryptophan, lipid, glycerophospholipid, amino acid, and carbohydrate metabolism, showed specific associations with sex and phenotype. Additionally, correlation analysis uncovered several differential metabolites that were significantly linked to mouse behaviors, with marked sex differences. Collectively, our results demonstrate a pronounced sexual dimorphism at the metabolic level in the hypothalamus and pituitary in response to chronic stress. This study provides a valuable molecular resource for further exploration of the interplay between sex and behavioral phenotypes within the dysregulation of the HPA axis that contributes to stress susceptibility and immune response, emphasizing the critical role of sex-specific metabolic mechanisms in anxiodepressive disorder.
Collapse
Affiliation(s)
- Lili Wang
- Institute of Neuroscience, School of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, China; Key Laboratory of Major Brain Disease and Aging Research (Ministry of Education), Chongqing Medical University, Chongqing 400016, China
| | - Bingtao Jiang
- Institute of Neuroscience, School of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, China; Key Laboratory of Major Brain Disease and Aging Research (Ministry of Education), Chongqing Medical University, Chongqing 400016, China
| | - Xunan Ji
- Institute of Neuroscience, School of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, China; Key Laboratory of Major Brain Disease and Aging Research (Ministry of Education), Chongqing Medical University, Chongqing 400016, China
| | - Jiaxin Tu
- Institute of Neuroscience, School of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, China; Key Laboratory of Major Brain Disease and Aging Research (Ministry of Education), Chongqing Medical University, Chongqing 400016, China
| | - Fengmei Lu
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Chen Yang
- Institute of Neuroscience, School of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, China; Key Laboratory of Major Brain Disease and Aging Research (Ministry of Education), Chongqing Medical University, Chongqing 400016, China
| | - Xianhui Zhong
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Lu Wang
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Xiao Cai
- Institute of Neuroscience, School of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, China; The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 611731, China; Key Laboratory of Major Brain Disease and Aging Research (Ministry of Education), Chongqing Medical University, Chongqing 400016, China
| | - Faping Yi
- Institute of Neuroscience, School of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, China; Key Laboratory of Major Brain Disease and Aging Research (Ministry of Education), Chongqing Medical University, Chongqing 400016, China
| | - Zongling He
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 611731, China.
| | - Liang Xie
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China.
| | - Jian Zhou
- Institute of Neuroscience, School of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, China; Key Laboratory of Major Brain Disease and Aging Research (Ministry of Education), Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
4
|
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
|
5
|
Sun D, Yang T, Wang M, Pang J, Li F. Sub-chronic exposure of hexaconazole may induce metabolic and neuropathic diseases: The evidence from gut microbiota. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2025; 210:106398. [PMID: 40262859 DOI: 10.1016/j.pestbp.2025.106398] [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: 07/05/2024] [Revised: 01/10/2025] [Accepted: 03/26/2025] [Indexed: 04/24/2025]
Abstract
The high-frequency detection and long persistence of hexaconazole (Hex) in agricultural products and environment poses potential risk to non-targeted organisms which should pay special attention to. Intestinal flora plays an important role in host health by prevention the occurrence of various diseases. Therefore, in this study, the disturbance of Hex on intestinal function and flora in rats had been studied at environmental related concentrations to evaluate the potential risk of Hex. Our results showed that Hex exposure induced serious oxidative stress and inflammation in intestinal tract. Meanwhile, it notably decreased the tight connectivity in colonic cell leading to the dysfunction of intestinal barrier. Moreover, 16sRNA gene sequencing showed that Hex exposure significantly disturbed the composition and structures of gut microbiota by decrease beneficial bacteria and increase pathogenic bacteria. Further, the metabolites and SCFAs that related to neuropathic and metabolic diseases in colonic contents were also significantly affected by Hex exposure. The pathways of membrane transport, replication and repair, lipid metabolism, and neurodegenerative diseases had been seriously interfered. The obtained results referred that Hex exposure may pose potential risk to metabolic system causing obesity, metabolic syndrome, and cardiovascular as well as nervous system inducing Parkinson's diseases, Alzheimer's diseases, and depression. Our study provided a new sight to study the mechanisms of Hex induced toxicity effects from the aspect of gut microbiota which could help for prevention the risk induced by Hex.
Collapse
Affiliation(s)
- Dali Sun
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang 561113, China.
| | - Tianming Yang
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang 561113, China
| | - Min Wang
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang 561113, China
| | - Junxiao Pang
- College of Food Science and Engineering, Guiyang University, Guiyang 550005, China
| | - Fumin Li
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang 561113, China
| |
Collapse
|
6
|
Biagini D, Di Franco C, Lazzarini G, Miragliotta V, Lomonaco T, Di Francesco F, Briganti A. Oxylipins as canine sepsis indicators in vivo and in ex vivo skin organ culture model. Sci Rep 2025; 15:12483. [PMID: 40216925 PMCID: PMC11992227 DOI: 10.1038/s41598-025-97460-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 04/04/2025] [Indexed: 04/14/2025] Open
Abstract
Sepsis, a life-threatening condition characterized by a dysregulated immune response to infection, remains a significant cause of mortality in both humans and veterinary patients. This study explores oxylipins as potential indicators of sepsis in dogs through in vivo plasma analysis and an ex vivo lipopolysaccharide (LPS)-treated skin organ culture model. By employing a robust analytical platform, 52 oxylipins and 4 polyunsaturated fatty acids were profiled in plasma and skin cultures. Results revealed distinct biochemical and morphological changes, with LPS triggering capillary vasodilation and time-dependent increases in pro-inflammatory mediators such as PGE2 and isoprostanes. Importantly, PGE2 exhibited consistent trends across both models, highlighting its potential as a diagnostic biomarker. This study underscores the utility of the skin organ culture model in mimicking early inflammatory events, offering novel insights into oxylipin dynamics during sepsis and their implications for disease resolution.
Collapse
Affiliation(s)
- Denise Biagini
- Department of Chemistry and Industrial Chemistry, University of Pisa, Pisa, Italy
| | - Chiara Di Franco
- Department of Veterinary Sciences, University of Pisa, Pisa, Italy
| | - Giulia Lazzarini
- Department of Veterinary Sciences, University of Pisa, Pisa, Italy
| | | | - Tommaso Lomonaco
- Department of Chemistry and Industrial Chemistry, University of Pisa, Pisa, Italy
| | - Fabio Di Francesco
- Department of Chemistry and Industrial Chemistry, University of Pisa, Pisa, Italy
- National Interuniversity Consortium of Materials Science and Technology (INSTM), Florence, Italy
| | - Angela Briganti
- Department of Veterinary Sciences, University of Pisa, Pisa, Italy
| |
Collapse
|
7
|
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
|
8
|
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
|
9
|
Yang Y, Liu Y, Williams TA, Gao M, Yan Y, Bao M, Tao J, Ma G, Wang M, Xia Z, Zhang Z, Yang T, Sun M. Metabolic phenotypes and fatty acid profiles associated with histopathology of primary aldosteronism. Hypertens Res 2025; 48:1363-1378. [PMID: 39939827 DOI: 10.1038/s41440-025-02143-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 12/30/2024] [Accepted: 01/24/2025] [Indexed: 02/14/2025]
Abstract
Primary aldosteronism (PA) caused by aldosterone hypersecretion is treated by adrenalectomy or medications. Histopathologic examination of resected adrenals reveals diverse histopathologic features. This study aimed to investigate the potential association of peripheral and adrenal tissue metabolic profiles with the histopathologic features of PA. The retrospective study included 105 surgically treated and 43 medically treated patients with PA. Adrenal specimens were categorized according to the HISTALDO (HISTopathology of primary ALDOsteronism) consensus. Peripheral and adrenal tissue metabolic profiles were assessed, including adiposity, adipokines and fatty acid abundances. The distinct fatty acid, arachidonic acid, was further functionally characterized. Surgically treated patients with classical histopathologic findings (n = 71) displayed lower body mass indexes, a lower prevalence of obesity, smaller waist circumference and visceral adipose tissue areas, and lower leptin concentrations compared with operated patients with the nonclassical histopathology (n = 34). No such differences were identified between the nonclassical histopathology group and medically treated group. Distinct concentrations of 18 out of 35 peripheral venous fatty acids, including arachidonic acid, were identified among the 3 groups. Further, accumulation of arachidonic acid was demonstrated in 4 aldosterone-producing adenomas compared with paired adjacent cortex possibly linked with suppressed peroxisomal beta-oxidation. Stimulation of human adrenocortical cells with arachidonic acid or peroxisomal beta-oxidation inhibitor caused 3.8-fold (P = 0.0050) and 1.7-fold (P = 0.0328) amplification of CYP11B2 expression, respectively, which were ablated by BAPTA-AM or KN93, and induced oxidative stress and apoptosis. Our findings show metabolic heterogeneity related to histopathology and support a role for arachidonic acid in PA pathophysiology.
Collapse
Affiliation(s)
- Yuhong Yang
- Department of Endocrinology, The First Affiliated Hospital With Nanjing Medical University, Nanjing, China
| | - Yuqing Liu
- Department of Endocrinology, The First Affiliated Hospital With Nanjing Medical University, Nanjing, China
| | - Tracy Ann Williams
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, LMU München, München, Germany
| | - Maoting Gao
- Department of Endocrinology, The First Affiliated Hospital With Nanjing Medical University, Nanjing, China
| | - Yutong Yan
- Department of Endocrinology, The First Affiliated Hospital With Nanjing Medical University, Nanjing, China
| | - Meiling Bao
- Department of Pathology, The First Affiliated Hospital With Nanjing Medical University, Nanjing, China
| | - Jun Tao
- Department of Urology, The First Affiliated Hospital With Nanjing Medical University, Nanjing, China
| | - Guodong Ma
- Department of Endocrinology, The First Affiliated Hospital With Nanjing Medical University, Nanjing, China
| | - Min Wang
- Department of Endocrinology, The First Affiliated Hospital With Nanjing Medical University, Nanjing, China
| | - Zhiqing Xia
- Department of Endocrinology, The First Affiliated Hospital With Nanjing Medical University, Nanjing, China
| | - Zhiheng Zhang
- Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China.
| | - Tao Yang
- Department of Endocrinology, The First Affiliated Hospital With Nanjing Medical University, Nanjing, China.
| | - Min Sun
- Department of Endocrinology, The First Affiliated Hospital With Nanjing Medical University, Nanjing, China.
| |
Collapse
|
10
|
Stepaniuk A, Sztolsztener K, Konstantynowicz-Nowicka K, Harasim-Symbor E, Bielawiec P, Chabowski A. The Identification of Novel Anti-Inflammatory Effects of Cannabigerol in the Kidney Tissue of Rats Subjected to a High-Fat High-Sucrose Diet. Int J Mol Sci 2025; 26:3114. [PMID: 40243749 PMCID: PMC11988375 DOI: 10.3390/ijms26073114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 03/21/2025] [Accepted: 03/25/2025] [Indexed: 04/18/2025] Open
Abstract
The inflammatory state is a significant factor associated with diabetic kidney disease (DKD), making it one of the significant causes of chronic kidney disease. Despite the availability of data, there is a lack of targeted treatment strategies for diabetes-related kidney disorders. The aim of our study was to determine the impact of cannabigerol (CBG) on lipid precursors for inflammatory mediators during DKD development. A six-week experiment was conducted on male Wistar rats fed standard (Control) or high-fat high-sucrose (HFHS) diets. For the last 14 days of the experiment (5th and 6th weeks), half of the rats from the Control and HFHS groups intragastrically received CBG solution. Gas-liquid chromatography (GLC) was used to measure the activities of n-6 and n-3 polyunsaturated fatty acid (PUFA) metabolic pathways and the concentrations of arachidonic acid (AA), eicosapentaenoic acid (EPA), and docosahexaenoic acid (DHA) in selected lipid fractions. Immunoblotting was performed to assess the expression of proteins involved in the regulation of the inflammatory state. A multiplex immunoassay kit was used to determine kidney toxicity biomarker levels. Our results revealed that CBG administration to rats fed an HFHS diet decreased n-6 PUFA biosynthetic pathway activity in phospholipid (PL) and triacylglycerol (TAG) and increased n-3 PUFA biosynthetic pathway activity in TAG and free fatty acid (FFA). We also observed a reduction in the AA concentration in PL, FFA, and diacylglycerol (DAG). CBG supplementation reduced the level of kidney damage biomarkers, such as osteopontin (OPN). Our observations confirm that CBG has potential anti-inflammatory properties and may be successfully used for further research to seek targeted therapies of inflammatory disorders, including diabetic kidney disease progression.
Collapse
Affiliation(s)
- Anna Stepaniuk
- Department of Physiology, Medical University of Bialystok, 15-089 Bialystok, Poland; (K.S.); (K.K.-N.); (E.H.-S.); (P.B.); (A.C.)
| | | | | | | | | | | |
Collapse
|
11
|
Wu L, Chen HY, Zhang JT, Yang RY, Wang ZB, Xue PS, Peng W, Li KX, Gao WH, Zeng PH. Chlorogenic acid induces hepatocellular carcinoma cell ferroptosis via PTGS2/AKR1C3/GPX4 axis-mediated reprogramming of arachidonic acid metabolism. World J Gastrointest Oncol 2025; 17:98844. [PMID: 40092947 PMCID: PMC11866246 DOI: 10.4251/wjgo.v17.i3.98844] [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: 07/07/2024] [Revised: 11/08/2024] [Accepted: 12/16/2024] [Indexed: 02/14/2025] Open
Abstract
BACKGROUND Ferroptosis is an iron-dependent programmed non-apoptotic cell death characterized by the accumulation of free iron ions and lipid peroxidation. It is associated with the inactivation of glutathione peroxidase (GPX) and the accumulation of lipid peroxides within cells. Ferroptosis is closely related to the occurrence and development of hepatocellular carcinoma (HCC). Chlorogenic acid (CGA), an important bioactive component found in 61 traditional Chinese medicines such as Eucommia ulmoides, has been extensively studied for its effects on various malignant tumors. However, the specific role and potential mechanism of CGA in HCC remain unclear. AIM To elucidate the anti-tumor characteristics and potential mechanisms of CGA in inducing ferroptosis in HCC cells. METHODS The effects of CGA on the proliferation, migration, and invasion of HCC cells were evaluated through in vitro experiments. Bioinformatics analysis combined with network pharmacology was used to study the potential targets and molecular mechanisms of CGA intervention in HCC ferroptosis. In vitro experiments were conducted to verify and explore the anti-HCC effects and mechanisms of CGA through the ferroptosis pathway. RESULTS In vitro experiments showed that CGA dose-dependently inhibited the proliferation, invasion, and migration of HCC cells. Bioinformatics analysis combined with network pharmacology revealed that the pathway of CGA intervention in HCC cell ferroptosis was mainly enriched in the prostaglandin endoperoxide synthase 2 (PTGS2)/aldo-keto reductase family 1 member C3 (AKR1C3)/GPX4 signaling pathway, which was associated with arachidonic acid. In vitro experiments further confirmed that CGA-induced ferroptosis in HCC cells was related to mitochondrial damage through the reprogramming of arachidonic acid metabolism by regulating the PTGS2/AKR1C3/GPX4 signaling pathway. CONCLUSION This study demonstrates that CGA inhibits HCC cell proliferation, migration, and invasion by inducing ferroptosis through the PTGS2/AKR1C3/GPX4 axis, suggesting its potential as a novel ferroptosis inducer or anti-HCC drug.
Collapse
Affiliation(s)
- Ling Wu
- College of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha 410208, Hunan Province, China
| | - Hong-Yao Chen
- College of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha 410208, Hunan Province, China
| | - Jing-Ting Zhang
- College of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha 410208, Hunan Province, China
| | - Ren-Yi Yang
- College of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha 410208, Hunan Province, China
| | - Zhi-Bin Wang
- College of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha 410208, Hunan Province, China
| | - Pei-Sen Xue
- College of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha 410208, Hunan Province, China
| | - Wei Peng
- Department of Oncology, Hunan Provincial Hospital of Integrated Traditional Chinese and Western Chinese, Changsha 410006, Hunan Province, China
| | - Ke-Xiong Li
- Department of Oncology, Hunan Provincial Hospital of Integrated Traditional Chinese and Western Chinese, Changsha 410006, Hunan Province, China
| | - Wen-Hui Gao
- College of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha 410208, Hunan Province, China
| | - Pu-Hua Zeng
- Cancer Research Institute of Hunan Academy of Traditional Chinese Medicine, Hunan Provincial Hospital of Integrated Traditional Chinese and Western Chinese, Changsha 410006, Hunan Province, China
| |
Collapse
|
12
|
Wang Y, Hsu P, Hu H, Lin F, Wei X. Role of arachidonic acid metabolism in osteosarcoma prognosis by integrating WGCNA and bioinformatics analysis. BMC Cancer 2025; 25:445. [PMID: 40075313 PMCID: PMC11905593 DOI: 10.1186/s12885-024-13278-3] [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: 08/01/2024] [Accepted: 12/02/2024] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND Osteosarcoma is a rare tumor with poor clinical outcomes. New therapeutic targets are urgently needed. Previous research indicates that genes abnormally expressed in osteosarcoma are significantly involved in the arachidonic acid (AA) metabolic pathway. However, the role of arachidonic acid metabolism-related genes (AAMRGs) in osteosarcoma prognosis remains unknown. METHODS Osteosarcoma samples from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases were classified into high-score and low-score groups based on AAMRGs scores obtained through ssGSEA analysis. The intersecting genes were identified from weighted gene co-expression network analysis (WGCNA), DEGs (osteosarcoma vs. normal) and DE-AAMRGs (high- vs. low-score). An AA metabolism predictive model of the five AAMRGs were established by Cox regression and the LASSO algorithm. Model performance was evaluated using Kaplan-Meier survival and receiver operating characteristic (ROC) curve analysis. In vitro experiments of the AA related biomarkers was validated. RESULTS Our study constructed an AAMRGs prognostic signature (CD36, CLDN11, STOM, EPYC, PANX3). K-M analysis indicated that patients in the low-risk group showed superior overall survival to high-risk group (p<0.05). ROC curves showed that all AUC values in the prognostic model exceeded 0.76. By ESTIMATE algorithms, we discovered that patients in high-risk groups had lower immune score, stromal score, and estimate score. Correlation analysis showed the strongest positive correlation between STOM and natural killer cells, and the highest negative association between PANX3 and central memory CD8 T cells. An AAMRGs prognostic signature was constructed for osteosarcoma prognosis. CONCLUSION The study suggested that a high level of AAMRGs might serve as a biomarker for poor prognosis in osteosarcoma and offers a potential explanation for the role of cyclooxygenase inhibitors in cancer. The five biomarkers (CD36, CLDN11, EPYC, PANX3, and STOM) were screened to construct an AAMRGs risk model with prognostic value, providing a new reference for the prognosis and treatment of osteosarcoma.
Collapse
Affiliation(s)
- Yaling Wang
- Department of Oncology, Shanghai Eighth People's Hospital, Shanghai, China
| | - Peichun Hsu
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haiyan Hu
- Shanghai Clinical Research Ward (SCRW), Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Feng Lin
- Department of Oncology, Shanghai Eighth People's Hospital, Shanghai, China.
| | - Xiaokang Wei
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
13
|
Fang X, Border JJ, Zhang H, Challagundla L, Kaur J, Hwang SH, Hammock BD, Fan F, Roman RJ. A Soluble Epoxide Hydrolase Inhibitor Improves Cerebrovascular Dysfunction, Neuroinflammation, Amyloid Burden, and Cognitive Impairments in the hAPP/PS1 TgF344-AD Rat Model of Alzheimer's Disease. Int J Mol Sci 2025; 26:2433. [PMID: 40141075 PMCID: PMC11942141 DOI: 10.3390/ijms26062433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 03/04/2025] [Accepted: 03/06/2025] [Indexed: 03/28/2025] Open
Abstract
Alzheimer's disease (AD) is an increasing global healthcare crisis with few effective treatments. The accumulation of amyloid plaques and hyper-phosphorylated tau are thought to underlie the pathogenesis of AD. However, current studies have recognized a prominent role of cerebrovascular dysfunction in AD. We recently reported that SNPs in soluble epoxide hydrolase (sEH) are linked to AD in human genetic studies and that long-term administration of an sEH inhibitor attenuated cerebral vascular and cognitive dysfunction in a rat model of AD. However, the mechanisms linking changes in cerebral vascular function and neuroprotective actions of sEH inhibitors in AD remain to be determined. This study investigated the effects of administration of an sEH inhibitor, 1-(1-Propanoylpiperidin-4-yl)-3-[4-(trifluoromethoxy)phenyl]urea (TPPU), on neurovascular coupling, blood-brain barrier (BBB) function, neuroinflammation, and cognitive dysfunction in an hAPP/PS1 TgF344-AD rat model of AD. We observed predominant β-amyloid accumulation in the brains of 9-10-month-old AD rats and that TPPU treatment for three months reduced amyloid burden. The functional hyperemic response to whisker stimulation was attenuated in AD rats, and TPPU normalized the response. The sEH inhibitor, TPPU, mitigated capillary rarefaction, BBB leakage, and activation of astrocytes and microglia in AD rats. TPPU increased the expression of pre- and post-synaptic proteins and reduced loss of hippocampal neurons and cognitive impairments in the AD rats, which was confirmed in a transcriptome and GO analysis. These results suggest that sEH inhibitors could be a novel therapeutic strategy for AD.
Collapse
Affiliation(s)
- Xing Fang
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS 39216, USA; (X.F.); (J.J.B.); (H.Z.)
| | - Jane J. Border
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS 39216, USA; (X.F.); (J.J.B.); (H.Z.)
| | - Huawei Zhang
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS 39216, USA; (X.F.); (J.J.B.); (H.Z.)
| | - Lavanya Challagundla
- Molecular and Genomics Facility, University of Mississippi Medical Center, Jackson, MS 39216, USA; (L.C.); (J.K.)
| | - Jasleen Kaur
- Molecular and Genomics Facility, University of Mississippi Medical Center, Jackson, MS 39216, USA; (L.C.); (J.K.)
| | - Sung Hee Hwang
- Entomology and Nematology and UC Davis Comprehensive Cancer Center, University of California Davis, Davis, CA 95616, USA; (S.H.H.); (B.D.H.)
| | - Bruce D. Hammock
- Entomology and Nematology and UC Davis Comprehensive Cancer Center, University of California Davis, Davis, CA 95616, USA; (S.H.H.); (B.D.H.)
| | - Fan Fan
- Physiology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA;
| | - Richard J. Roman
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS 39216, USA; (X.F.); (J.J.B.); (H.Z.)
| |
Collapse
|
14
|
Cui M, Zhang Y, Tang Y, Fan Q, Chen X, Li J, Qiao C, Chen X, Lin R, Yu X, Zhao C. Hepatotoxicity of Phytolacca acinosa Roxb mediated by phytolaccagenin via ferroptosis/PPAR/P53/arachidonic acid metabolism. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 138:156433. [PMID: 39892312 DOI: 10.1016/j.phymed.2025.156433] [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: 09/26/2024] [Revised: 01/18/2025] [Accepted: 01/25/2025] [Indexed: 02/03/2025]
Abstract
BACKGROUND The traditional Chinese medicine Phytolacca acinosa Roxb (PAR), known as Shanglu, possesses recognized therapeutic benefits against many diseases. PAR is also hepatotoxic, making it a major public health problem. However, the specific toxic substances and molecular mechanisms of PAR remain unclear. Therefore, appropriate animal models and methods are essential to confirm the toxic components and related mechanisms of PAR. METHODS L-02 cells and zebrafish larvae at 4 days post-fertilization (4 dpf) were used as models and treated with various concentrations of phytolaccagenin (Phy), esculentoside A (EsA), and esculentoside H (EsH). The hepatotoxicity of three samples was assessed based on liver phenotype, pathological assessments, and biochemical index in zebrafish and proliferative activity, apoptosis level, and biochemical index in L02 cells. The transcriptomic technique was used to explore the related signaling pathways and potential mechanisms in vitro and in zebrafish , and the findings were validated by RT-PCR. RESULTS The results of acute toxicity tests indicated that Phy exhibited substantially more severe hepatotoxicity than EsA, while EsH did not lead to any obvious toxic effects. Especially, under sublethal exposure ( CONCLUSION This study identified Phy as a key hepatotoxic component of PAR. Furthermore, using transcriptomic techniques, we preliminarily investigated the hepatotoxic mechanisms of Phy in vitro and in vivo. The results of the present study showed that Phy affects several signaling pathways, including those involved in lipid metabolism, oxidative stress, and apoptosis, finally leading to hepatotoxicity. These findings provide invaluable insights into the safe use of PAR in clinical settings.
Collapse
Affiliation(s)
- Muyao Cui
- Beijing University of Chinese Medicine, Beijing 100029, China; Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Yao Zhang
- Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yang Tang
- Beijing University of Chinese Medicine, Beijing 100029, China
| | - Qiqi Fan
- Beijing University of Chinese Medicine, Beijing 100029, China
| | - Xiaolu Chen
- Beijing University of Chinese Medicine, Beijing 100029, China; Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, Beijing 100102, China; Traditional Chinese Medicine Processing Technology Inheritance Base of National Administration of Traditional Chinese Medicine, China
| | - Jiaqi Li
- Beijing University of Chinese Medicine, Beijing 100029, China; Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, Beijing 100102, China; Traditional Chinese Medicine Processing Technology Inheritance Base of National Administration of Traditional Chinese Medicine, China
| | - Chuanqi Qiao
- Beijing University of Chinese Medicine, Beijing 100029, China; Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, Beijing 100102, China; Traditional Chinese Medicine Processing Technology Inheritance Base of National Administration of Traditional Chinese Medicine, China
| | - Xue Chen
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Ruichao Lin
- Beijing University of Chinese Medicine, Beijing 100029, China; Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, Beijing 100102, China; Traditional Chinese Medicine Processing Technology Inheritance Base of National Administration of Traditional Chinese Medicine, China.
| | - Xue Yu
- Beijing University of Chinese Medicine, Beijing 100029, China.
| | - Chongjun Zhao
- Beijing University of Chinese Medicine, Beijing 100029, China; Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, Beijing 100102, China; Traditional Chinese Medicine Processing Technology Inheritance Base of National Administration of Traditional Chinese Medicine, China.
| |
Collapse
|
15
|
Li X, Yang L, Zhu L, Sun J, Xu C, Sun L. Identifying potential biomarkers and molecular mechanisms related to arachidonic acid metabolism in vitiligo. Front Mol Biosci 2025; 12:1536477. [PMID: 40078960 PMCID: PMC11896865 DOI: 10.3389/fmolb.2025.1536477] [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: 11/29/2024] [Accepted: 01/30/2025] [Indexed: 03/14/2025] Open
Abstract
Background Numerous studies have reported that dysregulation of fatty acid metabolic pathways is associated with the pathogenesis of vitiligo, in which arachidonic acid metabolism (AAM) plays an important role. However, the molecular mechanisms of AAM in the pathogenesis of vitiligo have not been clarified. Therefore, we aimed to identify the biomarkers and molecular mechanisms associated with AAM in vitiligo using bioinformatics methods. Methods The GSE75819 and GSE65127 datasets were used in this study as the training and validation sets, respectively, along with 58 AAM-related genes (AAM-RGs). The differentially expressed genes (DEGs) between the lesional and control groups in the training set were identified through differential expression analysis. A biomarker-based nomogram was constructed to predict the risk of vitiligo. Results 15 overlapping candidate genes were obtained between the DEGs and AAM-RGs. Machine-learning algorithms were used to identify six key genes as PTGDS, PNPLA8, FAAH, ABHD12, PTGS1, and MGLL. In both the training and validation sets, PTGDS, PNPLA8, and MGLL. In both the training and validation sets, PTGDS, PNPLA8, and MGLL were regarded as biomarkers. A nomogram based on these biomarkers showed potential for predicting the risk of vitiligo. Functional enrichment, immune cell infiltration, and regulatory network analyses were used to elucidate the molecular mechanisms. Conclusion In conclusion, PTGDS, PNPLA8, and MGLL were implicated in AAM to influence the pathogenesis of vitiligo. These findings offer insights into vitiligo treatment, although further research is needed for a comprehensive understanding.
Collapse
Affiliation(s)
- Xiaoqing Li
- Shaanxi Provincial Key Laboratory of Infection and Immune Diseases, Shaanxi Provincial People’s Hospital, Xi’an, Shaanxi, China
- Department of Dermatology, Shaanxi Provincial People’s Hospital, Xi’an, Shaanxi, China
| | - Li Yang
- Department of Dermatology, Shaanxi Provincial People’s Hospital, Xi’an, Shaanxi, China
| | - Longfei Zhu
- Department of Dermatology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Jingying Sun
- Shaanxi Provincial Key Laboratory of Infection and Immune Diseases, Shaanxi Provincial People’s Hospital, Xi’an, Shaanxi, China
- Shaanxi Province Research Center of Cell Immunological Engineering and Technology, Shaanxi Provincial People’s Hospital, Xi’an, Shaanxi, China
| | - Cuixiang Xu
- Shaanxi Provincial Key Laboratory of Infection and Immune Diseases, Shaanxi Provincial People’s Hospital, Xi’an, Shaanxi, China
- Shaanxi Province Research Center of Cell Immunological Engineering and Technology, Shaanxi Provincial People’s Hospital, Xi’an, Shaanxi, China
| | - Lijun Sun
- Shaanxi Provincial Key Laboratory of Infection and Immune Diseases, Shaanxi Provincial People’s Hospital, Xi’an, Shaanxi, China
- Shaanxi Province Research Center of Cell Immunological Engineering and Technology, Shaanxi Provincial People’s Hospital, Xi’an, Shaanxi, China
| |
Collapse
|
16
|
Arshad R, Wan J, Ai T, Yin C, Qin Y, Qin R, Liu J, Liu H. A targeted reformulation of safflower oil: Enhancing anti-inflammatory potential and market competitiveness through ω3 enrichment. Food Res Int 2025; 203:115793. [PMID: 40022324 DOI: 10.1016/j.foodres.2025.115793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 01/06/2025] [Accepted: 01/18/2025] [Indexed: 03/03/2025]
Abstract
Safflower seed oil, rich in linoleic acid, has a high ω6/ω3 ratio due to its negligible ω3 content, which is potentially linked to inflammation and metabolic disorders. Despite its recognized health potential, excessive consumption of pure safflower oil can lead to adverse outcomes. To address a possible solution to solve this problem, this study optimized the ω6/ω3 ratio by blending safflower oil with ω3-rich flaxseed oil, and investigated whether the nutritional profile and health benefits of the ω3-supplemented safflower oil could be improved. Results have shown that in high-fat diet-fed mice, the optimized safflower oil significantly reduced body weight gain, fat mass, and improved glucose tolerance. The optimized safflower oil also improved the serum lipid profiles by lowering triglyceride, total cholesterol, and LDL-C levels, while reducing pro-inflammatory markers. Moreover, the adjusted ω6/ω3 ratios led to increased microbial diversity, a favorable Firmicutes/Bacteroidetes ratio, and enrichment of beneficial bacteria like Helicobacteraceae, while reducing pro-inflammatory bacteria such as Deferribacteraceae. These changes correlated with improved lipid metabolism and reduced fat accumulation. This study not only highlights a practical approach to improving safflower oil's health benefits but also provides a strategic direction for breeding programs to enhance its ω3 synthesis pathways, positioning safflower oil as a competitive and innovative alternative in the health food market.
Collapse
Affiliation(s)
- Rubab Arshad
- Hubei Provincial Key Laboratory for Protection and Application of Special Plant Germplasm in Wuling Area of China, College of Life Sciences, South-Central Minzu University, Wuhan 430070 China
| | - Jiawei Wan
- Hubei Provincial Key Laboratory for Protection and Application of Special Plant Germplasm in Wuling Area of China, College of Life Sciences, South-Central Minzu University, Wuhan 430070 China
| | - Tingyang Ai
- Hubei Provincial Key Laboratory for Protection and Application of Special Plant Germplasm in Wuling Area of China, College of Life Sciences, South-Central Minzu University, Wuhan 430070 China
| | - Cong Yin
- Hubei Provincial Key Laboratory for Protection and Application of Special Plant Germplasm in Wuling Area of China, College of Life Sciences, South-Central Minzu University, Wuhan 430070 China
| | - Yonghua Qin
- Hubei Provincial Key Laboratory for Protection and Application of Special Plant Germplasm in Wuling Area of China, College of Life Sciences, South-Central Minzu University, Wuhan 430070 China
| | - Rui Qin
- Hubei Provincial Key Laboratory for Protection and Application of Special Plant Germplasm in Wuling Area of China, College of Life Sciences, South-Central Minzu University, Wuhan 430070 China
| | - Jiao Liu
- Hubei Provincial Key Laboratory for Protection and Application of Special Plant Germplasm in Wuling Area of China, College of Life Sciences, South-Central Minzu University, Wuhan 430070 China.
| | - Hong Liu
- Hubei Provincial Key Laboratory for Protection and Application of Special Plant Germplasm in Wuling Area of China, College of Life Sciences, South-Central Minzu University, Wuhan 430070 China.
| |
Collapse
|
17
|
Wen F, Ling H, Ran R, Li X, Wang H, Liu Q, Li M, Yu T. LPCAT3 regulates the proliferation and metastasis of serous ovarian cancer by modulating arachidonic acid. Transl Oncol 2025; 52:102256. [PMID: 39733744 PMCID: PMC11743812 DOI: 10.1016/j.tranon.2024.102256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 11/30/2024] [Accepted: 12/23/2024] [Indexed: 12/31/2024] Open
Abstract
BACKGROUND Lysophosphatidylcholine acyltransferase 3 (LPCAT3) promotes ferroptosis through the incorporating polyunsaturated fatty acids into membrane phospholipids, however, its role in serous ovarian cancer remains unclear. Here explored cancer proliferation and metastasis after modulating LPCAP3. METHODS LPCAT3 protein in ovarian cancer tissues was detected using bioinformatic and immunohistoche mical assays. Cell behaviors were observed after up- or down-regulating LPCAT3. Lipid metabolites were determined, and then the pathway enrichment analysis was performed. RESULTS The expression level of LPCAT3 in serous ovarian cancer tissues was lower than that in other types of ovarian cancer, and high expression was associated with a longer survival time. Overexpressing LPCAT3 reduced cell proliferation, migration and invasion via enhancing ferroptosis and decreasing the survival signaling; these behaviors were enhanced in LPCAT3-downknocked cells, where a higher abundance of arachidonic acid was observed followed by up-regulation of the downstream survival signaling. In vivo, up-regulation of LPCAT3 decreased tumor growth, but down-regulation enhanced tumor growth and metastasis. CONCLUSIONS LPCAT3 modulated metabolism of arachidonic acid, thereby regulating ferroptosis and the survival signaling to determine cancer growth and metastasis.
Collapse
Affiliation(s)
- Fang Wen
- Laboratory of Obstetrics and Gynecology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Hongjian Ling
- Laboratory of Obstetrics and Gynecology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Rui Ran
- Laboratory of Obstetrics and Gynecology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Xinya Li
- Laboratory of Obstetrics and Gynecology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Houmei Wang
- Laboratory of Obstetrics and Gynecology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Qianfen Liu
- Women and Children's Hospital, Chongqing Medical University (Chongqing Health Center for Women and Children), China
| | - Min Li
- Laboratory of Obstetrics and Gynecology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China.
| | - Tinghe Yu
- Laboratory of Obstetrics and Gynecology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
18
|
Lu H, Zhang J, Wang Y. Identification of the Pharmacological Components and Its Targets of Sanghuang by Integration of Nontarget Metabolomics and Network Pharmacology Analysis. Biomed Chromatogr 2025; 39:e6066. [PMID: 39748251 DOI: 10.1002/bmc.6066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 10/09/2024] [Accepted: 12/06/2024] [Indexed: 01/04/2025]
Abstract
The objective of this study is to comprehensively to identify the core pharmacological components and their respective targets of three medicinal fungi Sanghuangs including Sanghuangporus vaninii (SV), Sanghuangporus lonicericola (SL), and Inonotus hispidus (IH). Metabolomics analysis indicated that a total of 495 and 660 differential metabolites were obtained in mycelium and fermentation broth samples among three Sanghuangs, respectively. The network pharmacology analysis showed that 6-[1]-ladderane hexanol, R-nostrenol, candidone, ellagic acid, and quercetin were the overlapping active ingredients of three Sanghuang species for diabetes mellitus, immune system disease, and neoplasm. Certonardosterol A, dalamid, and ethylene brassylate are unique active ingredients in SV, and certonardosterol K, kaempferide, and esculetin are unique active ingredients in SL. Asbestinine, neoandrographolide, isosakuranetin, and daucosterin are unique active ingredients in IH. Accordingly, the common core targets of active ingredients of the three Sanghuangs were ESR1, PIK3CA, and LYN. PRKCA, EGFR, and STAT3 were the unique targets of SV, SL, and IH, respectively. The primary active components and their respective targets, in addition to the component-target interaction of Sanghuangs that have been identified in the present study, provide a foundation for future research on the prevention and treatment of disease using Sanghuangs.
Collapse
Affiliation(s)
- Hengqian Lu
- School of Life Sciences, Anhui University, Hefei, Anhui, China
- Key Laboratory of Human Microenvironment and Precision Medicine of Anhui Higher Education Institutes, Anhui University, Hefei, Anhui, China
- Wuhu Dongyuan New Rural Co. Ltd., Wuhu, China
| | - Jintao Zhang
- School of Life Sciences, Anhui University, Hefei, Anhui, China
- Key Laboratory of Human Microenvironment and Precision Medicine of Anhui Higher Education Institutes, Anhui University, Hefei, Anhui, China
| | - Yongzhong Wang
- School of Life Sciences, Anhui University, Hefei, Anhui, China
- Key Laboratory of Human Microenvironment and Precision Medicine of Anhui Higher Education Institutes, Anhui University, Hefei, Anhui, China
- Anhui Province Joint Construction Discipline key Laboratory of Nanobody Technology, Hefei, China
| |
Collapse
|
19
|
Li X, Qin Y, Yue F, Lü X. Comprehensive Analysis of Fecal Microbiome and Metabolomics Uncovered dl-Norvaline-Ameliorated Obesity-Associated Disorders in High-Fat Diet-Fed Obese Mice by Targeting the Gut Microbiota. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:2381-2392. [PMID: 39808000 DOI: 10.1021/acs.jafc.4c06638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
Norvaline is a nonproteinogenic amino acid and an important food ingredient supplement for healthy food. In this study, dl-norvaline administration reduced body weight by more than 40% and improved glucose metabolism and energy metabolism in obese mice induced by a high-fat diet (HFD). Combination analysis of microbiome and metabolomics showed that dl-norvaline supplementation regulated gut bacteria structure, such as increasing beneficial bacteria (Mollicutes_RF39, Ruminococcaceae, Bacteroidaceae, Rikenellaceae, Lactobacillaceae, Clostridiaceae_1, uncultured_bacterium_f_Muribaculaceae, and Rikenellaceae_RC9_gut_group) and decreasing harmful bacteria (Fusobacteriia, Desulfovibrionales, Enterobacteriaceae, Burkholderiaceae, Helicobacteraceae, and Veillonellaceae) and modulated the metabolites involved in arachidonic acid metabolism, thus further promoting short-chain fatty acid production and improving gut barrier, thereby inflammatory responses and oxidative stress were ameliorated. In addition, the pseudogerm-free mouse model verified that dl-norvaline ameliorated obesity-associated disorders in HFD-fed obese mice by targeting gut microbiota. These results clarified that dl-norvaline may be promising for developing and innovating potential functional food products.
Collapse
Affiliation(s)
- Xin Li
- College of Food Science and Engineering, Northwest A&F University, Yangling, 712100 Shaanxi, China
- College of Life Sciences, Northwestern Polytechnical University, Xi'an, 710129 Shaanxi, China
| | - Yanting Qin
- College of Food Science and Engineering, Northwest A&F University, Yangling, 712100 Shaanxi, China
| | - Fangfang Yue
- College of Food Science and Engineering, Northwest A&F University, Yangling, 712100 Shaanxi, China
| | - Xin Lü
- College of Food Science and Engineering, Northwest A&F University, Yangling, 712100 Shaanxi, China
- Shaanxi Engineering Research Centre of Dairy Products Quality, Safety and Health, Yangling, 712100 Shaanxi, China
| |
Collapse
|
20
|
Ahmad S, Singh A, Akram W, Upadhyay A, Abrol GS. Algal lipids: A review on current status and future prospects in food processing. J Food Sci 2025; 90:e17618. [PMID: 39786345 DOI: 10.1111/1750-3841.17618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 11/30/2024] [Accepted: 12/06/2024] [Indexed: 01/12/2025]
Abstract
The consumer demand for functional foods derived from natural sources has been enhanced due to health-promoting effects. Algae are widely available globally as a sustainable source of proteins, lipids, and carbohydrates. Algal lipids are underexplored natural sources that exhibit several nutraceutical effects and applications in fortification, cosmetics, and pharmaceuticals. Both macro- and microalgae are composed of high-quality lipids. These latter involve polar lipids, nonpolar lipids, and essential fatty acids. Therefore, this review aimed to bring out knowledge on the chemistry of various lipids isolated and identified from micro- and macroalgae. Further, their extraction using traditional thermal (solid-liquid, and liquid-liquid) and advanced nonthermal (supercritical fluid, microwave-, ultrasound-, and enzyme-assisted) techniques has been explored. Along with this, bioactivities of algal lipids have been discussed. This study explored algal lipids in advancing sustainable food processing technologies that contribute positively to environmental sustainability and global health, in line with United Nations Sustainable Development GroupUnited Nations Sustainable Development Group UNSDGs.
Collapse
Affiliation(s)
- Sameer Ahmad
- Department of Bioengineering, Integral University, Lucknow, Uttar Pradesh, India
| | - Amit Singh
- Department of Postharvest Technology, Banda University of Agriculture & Technology, Banda, Uttar Pradesh, India
| | - Wasim Akram
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Ashutosh Upadhyay
- Department of Food Science and Technology, National Institute of Food Technology Entrepreneurship and Management Kundli, Sonipat, Haryana, India
| | - Ghan Shyam Abrol
- Department of Post-Harvest Technology, Rani Lakshmi Bai Central Agricultural University, Jhansi, Uttar Pradesh, India
| |
Collapse
|
21
|
Wang L, Ma JQ, Song LJ, Qu XP, Zhang Y, Fan HM, Wang C, Zheng LL, Gao GD, Qu Y, Shen LL, Liu B. Comprehensive multi-omics, behavioral and morphological analysis of the hazards of nano-plastics in mice with internal carotid artery occlusion. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 289:117711. [PMID: 39799923 DOI: 10.1016/j.ecoenv.2025.117711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 01/07/2025] [Accepted: 01/08/2025] [Indexed: 01/15/2025]
Abstract
Accumulation of nanoplastics (NPs) poses a severe threat to the homeostasis of the internal environment in patients with chronic diseases. The effects of NP contamination on health in chronically ill populations must urgently be elucidated. In this study, NPs injected via the tail vein were distributed in the brain and internal organs in a mouse model of chronic internal carotid occlusion. Mice with chronic internal carotid artery occlusion exposed to NPs showed behavioral abnormalities, such as depression and anxiety, thus indicating detrimental effects of NPs on the brain. Subsequently, we used proteomics and metabolomics to analyze the specific mechanisms underlying the damaging effects of NP deposition in the brain. The findings helped explain the differences in the underlying biochemical responses at the microscopic level in mice after NP exposure. The NPs not only accumulated in the brain and caused pathologic damage, but also contributed to accelerating atherosclerosis in the mouse model of internal carotid artery occlusion. This work confirms the risk of NPs in a model of internal carotid artery occlusion and elucidates the mechanism underlying this harm; moreover, it provides theoretical support for developing strategies to decrease microplastic intake in patients with internal carotid artery occlusion.
Collapse
Affiliation(s)
- Lu Wang
- Department of Neurosurgery, Tangdu Hospital, Airforce Military Medical University, Xi'an, Shaanxi, China.
| | - Jia-Qi Ma
- Department of Neurosurgery, Tangdu Hospital, Airforce Military Medical University, Xi'an, Shaanxi, China.
| | - Li-Jia Song
- Department of Pediatrics, Tangdu Hospital, Airforce Military Medical University, Xi'an, China.
| | - Xiao-Peng Qu
- Department of Neurosurgery, Tangdu Hospital, Airforce Military Medical University, Xi'an, Shaanxi, China.
| | - Yue Zhang
- Department of Neurosurgery, Tangdu Hospital, Airforce Military Medical University, Xi'an, Shaanxi, China.
| | - Hai-Ming Fan
- Key Laboratory of Synthetic and Natural Functional Molecule of the Ministry of Education, College of Chemistry and Materials Science, Northwest University, Xi'an 710127, China.
| | - Chao Wang
- Department of Neurosurgery, Tangdu Hospital, Airforce Military Medical University, Xi'an, Shaanxi, China.
| | - Long-Long Zheng
- Department of Neurosurgery, Tangdu Hospital, Airforce Military Medical University, Xi'an, Shaanxi, China. zhenglong--
| | - Guo-Dong Gao
- Department of Neurosurgery, Tangdu Hospital, Airforce Military Medical University, Xi'an, Shaanxi, China.
| | - Yan Qu
- Department of Neurosurgery, Tangdu Hospital, Airforce Military Medical University, Xi'an, Shaanxi, China.
| | - Liang-Liang Shen
- Department of Biochemistry and Molecular Biology, Basic Medical Science Academy, Airforce Military Medical University, Xi'an, China.
| | - Bei Liu
- Department of Neurosurgery, Tangdu Hospital, Airforce Military Medical University, Xi'an, Shaanxi, China.
| |
Collapse
|
22
|
Vasudevan G, Ramachandran K, Tangavel C, Nayagam SM, Gopalakrishnan C, Muthurajan R, Sri Vijay Anand KS, Rajasekaran S. "Elucidating the immunomodulatory role of endocannabinoids in intervertebral disc degeneration". EUROPEAN SPINE JOURNAL : OFFICIAL PUBLICATION OF THE EUROPEAN SPINE SOCIETY, THE EUROPEAN SPINAL DEFORMITY SOCIETY, AND THE EUROPEAN SECTION OF THE CERVICAL SPINE RESEARCH SOCIETY 2025; 34:308-315. [PMID: 39542877 DOI: 10.1007/s00586-024-08550-w] [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: 07/23/2024] [Revised: 07/23/2024] [Accepted: 10/27/2024] [Indexed: 11/17/2024]
Abstract
PURPOSE The endocannabinoid system (ECS) has been well-established to play a crucial role in the regulation of several physiological processes as well as many inflammatory disease conditions. However, its role in intervertebral disc degeneration has been least explored. We aim to investigate the immunomodulatory role of endocannabinoids in regulating IVD health. METHODS The study population included 20 healthy volunteers (controls) and 40 patients with disc degeneration (disease group) (20 Modic and 20 Non Modic). 16S metagenome sequencing of the V3-V4 region was performed for the DNA extracted from NP tissue samples of both control and disease groups. Sequencing was carried out using the Novaseq 6000 platform using 250 bp paired-end chemistry. A global metabolic profile was obtained using the uHPLC system coupled with Q Exactive Plus Hybrid Quadrupole-Orbitrap mass spectrometer. RESULTS Our study revealed a higher prevalence of gram-negative bacteria, particularly opportunistic pathogens like Pseudomonas, in diseased discs (71-81%) compared to healthy controls (54%). Further investigation using metabolomics identified significant changes in the lipid profiles of diseased discs. We found that the signalling molecules of the ECS, 2-arachidonylglycerol (2-AG) and N-arachidonoylethanolamine (AEA), were significantly lower in diseased discs compared to controls (Log2FC -2.62 for 2-AG and -3.15 for AEA). Conversely, pro-inflammatory metabolites like LTA4, HPETE, HETE, and Prostaglandin G2 were elevated in diseased discs, with a Log2 fold increase greater than 2.5. CONCLUSION The study reveals that the endocannabinoid metabolites (2-AG and AEA) of the ECS could be a significant molecule influencing susceptibility to infection and inflammation within the intervertebral discs, which could be a potential target for improving disc health. LEVEL OF EVIDENCE Diagnostic: individual cross-sectional studies with consistently applied reference standard and blinding.
Collapse
Affiliation(s)
- Gowdaman Vasudevan
- Ganga Research Centre, SF No.442, NGGO Colony Post, Vattamalaipalayam, Coimbatore, TamilNadu, 641022, India
| | - Karthik Ramachandran
- Department of Spine Surgery, Ganga Hospital, 313, Mettupalayam Road, Coimbatore, India
| | - Chitraa Tangavel
- Ganga Research Centre, SF No.442, NGGO Colony Post, Vattamalaipalayam, Coimbatore, TamilNadu, 641022, India
| | - Sharon Miracle Nayagam
- Ganga Research Centre, SF No.442, NGGO Colony Post, Vattamalaipalayam, Coimbatore, TamilNadu, 641022, India
| | - Chellappa Gopalakrishnan
- Ganga Research Centre, SF No.442, NGGO Colony Post, Vattamalaipalayam, Coimbatore, TamilNadu, 641022, India
| | - Raveendran Muthurajan
- Department of Plant Biotechnology, Tamil Nadu Agricultural University, Lawley Road, Coimbatore, India
| | - K S Sri Vijay Anand
- Department of Spine Surgery, Ganga Hospital, 313, Mettupalayam Road, Coimbatore, India
| | | |
Collapse
|
23
|
Yang Y, Liu Y, Cheng Y, He H, Liang A, Pan Z, Liu Y, Chen Z. Multi-omics and experimental analysis unveil the key components in Scutellaria baicalensis Georgi to alleviate hepatic fibrosis via regulating cPLA2-mediated arachidonic acid metabolism. J Transl Med 2024; 22:1138. [PMID: 39716274 DOI: 10.1186/s12967-024-05955-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 12/07/2024] [Indexed: 12/25/2024] Open
Abstract
BACKGROUND Scutellaria baicalensis Georgi, a traditional Chinese herb, is known for its various biological effects, including antibacterial, anti-inflammatory, antioxidative, and antitumor properties. However, the function and mechanisms of methanol extract of Scutellaria baicalensis Georgi (MESB) in treating hepatic fibrosis remain unclear. METHODS This study utilized a CCl4-induced mouse model of hepatic fibrosis to assess the effects of MESB through histopathological analysis and serum tests. The anti-fibrosis mechanism of MESB was investigated using qPCR, Western blotting, RNA interference, proteomics, and metabolomics. Spatial metabolomics identified key components of MESB in liver tissue, while molecular docking determined their targets. RESULTS Treatment with MESB alleviated hepatic pathological changes and reversed hepatic fibrosis in the CCl4-induced models, as evidenced by decreased collagen fibers deposition, reduced expression of hepatic fibrosis markers COL1A1, FN, and PAI-1, and lowered serum levels of AST and ALT. In vitro, MESB inhibited the proliferation of LX-2 cells and the expression of hepatic fibrosis markers. Furthermore, MESB intervention modulated various pathways, particularly those involved in metabolic pathways. Subsequent metabolomics analysis demonstrated that MESB disrupted glycerophospholipid metabolism and suppressed arachidonic acid metabolism. MESB downregulated the expression of cPLA2 in LX-2 cells, leading to decreased production of arachidonic acid and its downstream inflammatory mediators. Meanwhile, MESB inhibited the expression of cPLA2 and its downstream NF-κB pathway in the liver tissues of models induced by CCl4. Additionally, silencing cPLA2 markedly reduced the expressions of COL1A1, FN, and PAI-1. Spatial metabolomics analysis confirmed the penetration of baicalein, wogonin and wogonoside into liver tissue. Further results indicated that baicalein and wogonin inhibited the expression of cPLA2, while baicalin and wogonoside do not exhibit this effect. Moreover, molecular docking suggested that baicalein and wogonin possess the potential to directly interact with cPLA2. CONCLUSION This study reveals that MESB is crucial in preventing hepatic fibrosis via the cPLA2-mediated arachidonic acid metabolic pathway, highlighting its key active components as potential drugs for fibrosis treatment.
Collapse
Affiliation(s)
- Yunheng Yang
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Yi Liu
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Yujie Cheng
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Honglin He
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Ailing Liang
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Zheng Pan
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Yuanyuan Liu
- Department of Radiological Medicine, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, China.
| | - Zhiwei Chen
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing, 400016, China.
- College of Traditional Chinese Medicine, Chongqing University of Chinese Medicine, Chongqing, 402760, China.
| |
Collapse
|
24
|
Correa-Navarro VA, Romo-Morales GDC, Sánchez-Palafox JE, Rodríguez-Ríos D, Molina-Torres J, Ramírez-Chávez E, Zaina S, Lund G. A Survey of Fatty Acid Content of the Male Reproductive System in Mice Supplemented With Arachidonic Acid. J Lipids 2024; 2024:3351340. [PMID: 39734583 PMCID: PMC11671656 DOI: 10.1155/jl/3351340] [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: 07/29/2024] [Accepted: 11/15/2024] [Indexed: 12/31/2024] Open
Abstract
Paternal exposure to high-fat diets or individual fatty acids (FAs) including arachidonic acid (AA) modifies progeny traits by poorly understood mechanisms. Specific male reproductive system FAs may be involved in paternal inheritance, as they can modify a range of cellular components, including the epigenome. Our objective was to determine FAs in compartments of the male reproductive system that potentially affect ejaculate composition-right and left testicular interstitial fluid (TIF), vesicular gland fluid (VGF), and epididymal adipose tissue (EAT)-in mice exposed to AA or vehicle daily for 10 days (n = 9-10/group). Whole blood (WB) and interscapular brown adipose tissue (IBAT) FA profiles were used as reference. AA significantly affected only VGF FAs relative to vehicle, that is, increased and decreased levels of arachidic and docosahexaenoic acid, respectively, versus vehicle (0.28% ± 0.01% and 0.23% ± 0.03%, respectively, p = 0.049, and 2.42% ± 0.47% and 3.00% ± 0.58%, respectively, p = 0.041). AA affected distinct FAs in WB. Additionally, we uncovered AA-dependent and AA-independent FA laterality. Myristic acid was higher in AA-exposed left versus right TIF (0.68% ± 0.35% and 0.60% ± 0.11%, respectively, p = 0.004). Right TIF contained higher oleic and linoleic acid and lower stearic acid than left TIF (29.01% ± 3.07% and 24.00% ± 2.18%, respectively, p = 0.005; 9.14% ± 1.88% and 7.05% ± 1.36%, respectively, p = 0.005; and 21.90% ± 2.92% and 26.01% ± 2.46%, respectively, p = 0.036), irrespective of exposure to AA. The TIF oleic/stearic acid ratio suggested higher Stearoyl-CoA Desaturase 1 activity in the right versus the left testis (1.35 ± 0.32 and 1.00 ± 0.17, respectively, p = 1.0 × 10-4). Multitissue comparisons revealed that TIF and VGF FA profiles were distinct from WB, EAT, or IBAT counterparts, suggesting tissue-specific FA fingerprints. In conclusion, AA modulated selected VGF long-chain FAs that may impact on uterine inflammation and subsequent embryonic development. AA altered local FA synthesis or selective uptake, rather than eliciting passive uptake from WB. Additionally, we uncover a significant laterality of testis FAs that may result in asymmetric sperm cell phenotypes.
Collapse
Affiliation(s)
- Viridiana Abigail Correa-Navarro
- Department of Medical Sciences, Division of Health Sciences, Leon Campus, University of Guanajuato, 20 de Enero 929, Leon, Guanajuato, Mexico
| | - Gloria del Carmen Romo-Morales
- Department of Medical Sciences, Division of Health Sciences, Leon Campus, University of Guanajuato, 20 de Enero 929, Leon, Guanajuato, Mexico
| | - Jaime Eduardo Sánchez-Palafox
- Department of Medical Sciences, Division of Health Sciences, Leon Campus, University of Guanajuato, 20 de Enero 929, Leon, Guanajuato, Mexico
| | - Dalia Rodríguez-Ríos
- Department of Genetic Engineering, CINVESTAV Irapuato Unit, Km 9.6 Libramiento Norte, Carretera Irapuato-León, Irapuato, Guanajuato 36824, Mexico
| | - Jorge Molina-Torres
- Department of Biotechnology and Biochemistry, CINVESTAV Irapuato Unit, Km 9.6 Libramiento Norte, Carretera Irapuato-León, Irapuato, Guanajuato 36824, Mexico
| | - Enrique Ramírez-Chávez
- Department of Biotechnology and Biochemistry, CINVESTAV Irapuato Unit, Km 9.6 Libramiento Norte, Carretera Irapuato-León, Irapuato, Guanajuato 36824, Mexico
| | - Silvio Zaina
- Department of Medical Sciences, Division of Health Sciences, Leon Campus, University of Guanajuato, 20 de Enero 929, Leon, Guanajuato, Mexico
| | - Gertrud Lund
- Department of Genetic Engineering, CINVESTAV Irapuato Unit, Km 9.6 Libramiento Norte, Carretera Irapuato-León, Irapuato, Guanajuato 36824, Mexico
| |
Collapse
|
25
|
Ergül AG, Jordan PM, Dahlke P, Bal NB, Olğaç A, Uludağ O, Werz O, Çalışkan B, Banoglu E. Novel Benzimidazole Derivatives as Potent Inhibitors of Microsomal Prostaglandin E 2 Synthase 1 for the Potential Treatment of Inflammation, Pain, and Fever. J Med Chem 2024; 67:21143-21162. [PMID: 39622054 DOI: 10.1021/acs.jmedchem.4c01883] [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: 12/13/2024]
Abstract
Microsomal prostaglandin E2 synthase 1 (mPGES-1) is a promising target for treating inflammatory diseases and pain. This study introduces a novel series of benzimidazoles, with the most potent analogs exhibiting IC50 values of 0.27-7.0 nM in a cell-free assay for prostaglandin (PG)E2 production. Compound 44 (AGU654) demonstrated remarkable selectivity for mPGES-1 (IC50 = 2.9 nM) over COX-1, COX-2, 5-LOX, and FLAP, along with excellent bioavailability. Metabololipidomics analysis with activated human monocyte-derived macrophages and human whole blood revealed that AGU654 selectively suppresses PGE2 production triggered by bacterial exotoxins while sparing other prostaglandins. Furthermore, in vivo studies showed that AGU654 significantly alleviated fever, inflammation, and inflammatory pain in preclinical guinea pig models, suggesting that it could be an effective strategy for managing inflammatory diseases. In conclusion, these benzimidazole derivatives warrant further exploration into new and alternative analogs, potentially uncovering novel compounds with a favorable pharmacological profile possessing significant anti-inflammatory and analgesic properties.
Collapse
Affiliation(s)
- Azize Gizem Ergül
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Gazi University, 06560 Ankara, Turkey
| | - Paul M Jordan
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Philosophenweg 14, D-7743 Jena, Germany
| | - Philipp Dahlke
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Philosophenweg 14, D-7743 Jena, Germany
| | - Nur Banu Bal
- Department of Pharmacology, Faculty of Pharmacy, Gazi University, 06560 Ankara, Turkey
| | - Abdurrahman Olğaç
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Gazi University, 06560 Ankara, Turkey
| | - Orhan Uludağ
- Department of Pharmacology, Faculty of Pharmacy, Gazi University, 06560 Ankara, Turkey
| | - Oliver Werz
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Philosophenweg 14, D-7743 Jena, Germany
| | - Burcu Çalışkan
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Gazi University, 06560 Ankara, Turkey
| | - Erden Banoglu
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Gazi University, 06560 Ankara, Turkey
| |
Collapse
|
26
|
Dai S, Zou L, Wang Q. Toxicity of organophosphate flame retardant in marine rotifers: Evidence from the population, individual, biochemical and molecular levels. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 955:177256. [PMID: 39477105 DOI: 10.1016/j.scitotenv.2024.177256] [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: 08/19/2024] [Revised: 10/25/2024] [Accepted: 10/25/2024] [Indexed: 11/03/2024]
Abstract
Tris (1-chloro-2-propyl) phosphoric acid (TCPP), a widely used organophosphate flame retardant, has been detected in various aquatic environments due to its extensive industrial application. TCPP is well-known to negatively impact large aquatic organisms. However, the effects of TCPP on zooplankton remain poorly understood. This study explored the ecological risk of TCPP in low-trophic marine organisms by evaluating the marine rotifer Brachionus plicatilis at the molecular, biochemical, individual, and population levels after exposure to TCPP concentrations of 14.79, 44.37, and 73.94 μM. Results showed that exposure to TCPP inhibited body size, feeding behavior, life expectancy, generation time, net reproductive rate, reproduction rate, and population growth rate of rotifers, thus impairing their growth, survival, reproduction, and population expansion. Environmental concentrations surpassing 0.031 μM and 0.23 μM adversely impact rotifer reproduction and survival, respectively. Biochemically, TCPP induced oxidative stress, increased amylase activity, decreased lipase activity, and total protein content. Transcriptome analysis revealed that TCPP could induce abnormal mitochondrial function, impaired energy metabolism, programmed cell death by generating excessive reactive oxygen species, and affect cellular DNA replication. Results indicate that TCPP disrupts homeostasis in rotifers by inducing oxidative stress, significantly suppressing individual and population parameters. These findings provide critical insights for assessing the ecological risk posed by TCPP to zooplankton and the stability of aquatic ecosystems.
Collapse
Affiliation(s)
- Shiyu Dai
- Department of Ecology, State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, 510632, China
| | - Ligong Zou
- Department of Ecology, State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, 510632, China
| | - Qing Wang
- Department of Ecology, State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
27
|
Werner R, Carnazza M, Li XM, Yang N. Effect of Small-Molecule Natural Compounds on Pathologic Mast Cell/Basophil Activation in Allergic Diseases. Cells 2024; 13:1994. [PMID: 39682741 PMCID: PMC11639848 DOI: 10.3390/cells13231994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 10/04/2024] [Accepted: 10/08/2024] [Indexed: 12/18/2024] Open
Abstract
Pathologic mast cells and basophils, key effector cells in allergic reactions, play pivotal roles in initiating and perpetuating IgE-mediated allergic responses. Conventional therapies for allergies have limitations, prompting exploration into alternative approaches such as small-molecule natural compounds derived from botanical sources. This review synthesizes the existing literature on the effects of these compounds on pathologic mast cells and basophils, highlighting their potential in allergy management, and utilizes the PubMed database for literature acquisition, employing keyword-based searches to identify relevant peer-reviewed sources. Additionally, mechanistic insights were evaluated to contextualize how small-molecule natural compounds can inhibit mast cell/basophil activation, degranulation, and signaling pathways crucial for IgE-mediated allergic reactions. Small-molecule natural compounds exhibit promising anti-allergic effects, yet despite these findings, challenges persist in the development and translation of natural compound-based therapies, including bioavailability and standardization issues. Future research directions include optimizing dosing regimens, exploring synergistic effects with existing therapies, and employing systems pharmacology approaches for a holistic understanding of their mechanisms of action. By harnessing the therapeutic potential of small-molecule natural compounds, effective treatments for allergic diseases may be realized, offering hope for individuals with allergies.
Collapse
Affiliation(s)
- Robert Werner
- Division of R&D, General Nutraceutical Technology LLC, Elmsford, NY 10523, USA; (R.W.); (M.C.)
| | - Michelle Carnazza
- Division of R&D, General Nutraceutical Technology LLC, Elmsford, NY 10523, USA; (R.W.); (M.C.)
| | - Xiu-Min Li
- Department of Pathology, Microbiology & Immunology, New York Medical College, Valhalla, NY 10595, USA;
- Department of Otolaryngology, School of Medicine, New York Medical College, Valhalla, NY 10595, USA
- Department of Dermatology, New York Medical College, Valhalla, NY 10595, USA
| | - Nan Yang
- Division of R&D, General Nutraceutical Technology LLC, Elmsford, NY 10523, USA; (R.W.); (M.C.)
| |
Collapse
|
28
|
Yao D, Shen C, Zhang X, Tang J, Yu J, Tu M, Panpipat W, Chaijan M, Zhang H, Xu X, Liu Y, Cheong LZ. Untargeted metabolomics study of mature human milk from women with and without gestational diabetes mellitus. Food Chem 2024; 460:140663. [PMID: 39142199 DOI: 10.1016/j.foodchem.2024.140663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 07/25/2024] [Accepted: 07/25/2024] [Indexed: 08/16/2024]
Abstract
Gestational diabetes mellitus (GDM) is a prevalent metabolic disorder during pregnancy that alters the metabolites in human milk. Integrated Gas Chromatography-Mass Spectrometry (GC-MS) and Liquid Chromatography-Mass Spectrometry (LC-MS) were employed for comprehensive identification and comparison of metabolites in mature human milk (MHM) from women with and without GDM. A total of 268 differentially expressed metabolites (DEMs) were identified. Among these, linoleic acid, arachidonic acid, 9R-HODE and L-glutamic acid were significantly elevated and 12,13-DHOME was significantly decreased in MHM of women with GDM. These metabolites are significantly enriched in linoleic acid metabolism, fatty acid biosynthesis, galactose metabolism and ABC transporters pathways. Disorders in these metabolic pathways are associated with insulin resistance and poor glucose metabolism indicating these conditions may persist postpartum.
Collapse
Affiliation(s)
- Dan Yao
- Zhejiang-Malaysia Joint Research Laboratory for Agricultural Product Processing and Nutrition, Key Laboratory of Animal Protein Food Deep Processing Technology of Zhejiang Province, College of Food Science and Engineering, Ningbo University, Ningbo 315800, China
| | - Cai Shen
- School of Agriculture and Food, Faculty of Science, University of Melbourne, 3010, Australia
| | - Xinghe Zhang
- School of Agriculture and Food, Faculty of Science, University of Melbourne, 3010, Australia
| | - Jiayue Tang
- Zhejiang-Malaysia Joint Research Laboratory for Agricultural Product Processing and Nutrition, Key Laboratory of Animal Protein Food Deep Processing Technology of Zhejiang Province, College of Food Science and Engineering, Ningbo University, Ningbo 315800, China
| | - Jingwen Yu
- Zhejiang-Malaysia Joint Research Laboratory for Agricultural Product Processing and Nutrition, Key Laboratory of Animal Protein Food Deep Processing Technology of Zhejiang Province, College of Food Science and Engineering, Ningbo University, Ningbo 315800, China
| | - Maolin Tu
- Zhejiang-Malaysia Joint Research Laboratory for Agricultural Product Processing and Nutrition, Key Laboratory of Animal Protein Food Deep Processing Technology of Zhejiang Province, College of Food Science and Engineering, Ningbo University, Ningbo 315800, China
| | - Worawan Panpipat
- School of Agricultural Technology and Food Industry, Walailak University, 80161, 222 Thaiburi, Thasala District, Nakhonsrithammarat, Thailand
| | - Manat Chaijan
- School of Agricultural Technology and Food Industry, Walailak University, 80161, 222 Thaiburi, Thasala District, Nakhonsrithammarat, Thailand
| | - Hong Zhang
- Wilmar (Shanghai) Biotechnology Research and Development Center Co Ltd., No.118 Gaodong Rd., Pudong New District, Shanghai 200137, China
| | - Xuebing Xu
- Wilmar (Shanghai) Biotechnology Research and Development Center Co Ltd., No.118 Gaodong Rd., Pudong New District, Shanghai 200137, China
| | - Yanan Liu
- Zhejiang-Malaysia Joint Research Laboratory for Agricultural Product Processing and Nutrition, Key Laboratory of Animal Protein Food Deep Processing Technology of Zhejiang Province, College of Food Science and Engineering, Ningbo University, Ningbo 315800, China
| | - Ling-Zhi Cheong
- School of Agriculture and Food, Faculty of Science, University of Melbourne, 3010, Australia
| |
Collapse
|
29
|
Pang Q, Qi X, Chi Y, Jiajue R, Zhang L, Cui L, Wang O, Li M, Xing X, Jiang Y, Gong Y, Xia W. Targeting Metabolomics in Primary Hypertrophic Osteoarthropathy: Uncovering Novel Insights into Disease Pathogenesis. J Clin Endocrinol Metab 2024:dgae737. [PMID: 39607761 DOI: 10.1210/clinem/dgae737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Indexed: 11/29/2024]
Abstract
CONTEXT Primary hypertrophic osteoarthropathy (PHO) is a rare genetic disorder characterized by skeletal and skin abnormalities. Genetic defects in prostaglandin E2 (PGE2) metabolism are known to cause PHO. However, the global impact and clinical significance of eicosanoids and oxylipins beyond PGE2 remain to be elucidated. OBJECTIVE This study aimed to investigate oxylipin networks in PHO, including the 2 subtypes, PHOAR1 and PHOAR2, and examine their associations with clinical characteristics. METHODS We conducted a targeted metabolomic study involving 16 patients with PHO and 16 age- and sex-matched healthy controls. Serum samples were collected at the time of diagnosis. Metabolites were quantified using ultra-high-performance liquid chromatography-tandem mass spectrometry. RESULTS Laboratory analyses confirmed elevated levels of PGE2 in patients with PHO, consistent with the established pathogenesis. About 60 oxidized lipid metabolites were identified, with 19 differentially expressed in PHO. Besides the COX/PGE2 pathway, the lipoxygenase-mediated pathway was also involved in PHO. The metabolites 5-OxoETE, 15-OxoETE, 8S,15S-DiHETE, PGE2, 11β-PGE2, PGB2, LTB4, and LTE4 were significantly altered. Correlation analyses revealed associations between oxylipin metabolites and clinical features, including bone microarchitecture. Notably, the study highlighted differences in the oxylipin metabolite profiles between patients with PHOAR1 and patients with PHOAR2, suggesting distinct metabolic signatures for each subtype. CONCLUSION Our study indicated a significant perturbation in oxylipin metabolism among patients with PHO, with distinct metabolic signatures observed between PHOAR1 and PHOAR2. The disruption extended beyond the metabolism of PGE2. It encompassed a broader alteration across the polyunsaturated fatty acid metabolism spectrum, including various eicosanoids and oxylipins. Our work provided a comprehensive understanding of the pathogenesis of PHO, and underscored the potential for subtype-specific therapeutic interventions.
Collapse
Affiliation(s)
- Qianqian Pang
- Department of Endocrinology, Key Laboratory of Endocrinology, National Commission of Health, State Key Laboratory for Complex, Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science, Beijing 100730, China
| | - Xuan Qi
- Department of Endocrinology, Key Laboratory of Endocrinology, National Commission of Health, State Key Laboratory for Complex, Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science, Beijing 100730, China
| | - Yue Chi
- Department of Endocrinology, Key Laboratory of Endocrinology, National Commission of Health, State Key Laboratory for Complex, Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science, Beijing 100730, China
| | - Ruizhi Jiajue
- Department of Endocrinology, Key Laboratory of Endocrinology, National Commission of Health, State Key Laboratory for Complex, Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science, Beijing 100730, China
| | - Li Zhang
- Department of Endocrinology, Key Laboratory of Endocrinology, National Commission of Health, State Key Laboratory for Complex, Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science, Beijing 100730, China
| | - Lijia Cui
- Department of Endocrinology, Key Laboratory of Endocrinology, National Commission of Health, State Key Laboratory for Complex, Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science, Beijing 100730, China
| | - Ou Wang
- Department of Endocrinology, Key Laboratory of Endocrinology, National Commission of Health, State Key Laboratory for Complex, Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science, Beijing 100730, China
| | - Mei Li
- Department of Endocrinology, Key Laboratory of Endocrinology, National Commission of Health, State Key Laboratory for Complex, Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science, Beijing 100730, China
| | - Xiaoping Xing
- Department of Endocrinology, Key Laboratory of Endocrinology, National Commission of Health, State Key Laboratory for Complex, Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science, Beijing 100730, China
| | - Yan Jiang
- Department of Endocrinology, Key Laboratory of Endocrinology, National Commission of Health, State Key Laboratory for Complex, Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science, Beijing 100730, China
| | - Yiyi Gong
- Department of Endocrinology, Key Laboratory of Endocrinology, National Commission of Health, State Key Laboratory for Complex, Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science, Beijing 100730, China
- Center for Biomarker Discovery and Validation, National Infrastructures for Translational Medicine, Institute of Clinical Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Weibo Xia
- Department of Endocrinology, Key Laboratory of Endocrinology, National Commission of Health, State Key Laboratory for Complex, Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science, Beijing 100730, China
| |
Collapse
|
30
|
Haj-Yahya F, Steinberg D, Sionov RV. Trans, Trans-Farnesol Enhances the Anti-Bacterial and Anti-Biofilm Effect of Arachidonic Acid on the Cariogenic Bacteria Streptococcus mutans and Streptococcus sobrinus. Int J Mol Sci 2024; 25:11770. [PMID: 39519322 PMCID: PMC11546208 DOI: 10.3390/ijms252111770] [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: 10/13/2024] [Revised: 10/26/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Streptococcus mutans and Streptococcus sobrinus are Gram-positive bacteria involved in the development of dental caries, as they are able to form biofilms on tooth enamel, ferment sugars into acids, and survive under acidic conditions. This ultimately leads to a local lowering of the pH value on the tooth surface, which causes enamel cavities. HYPOTHESIS One measure to reduce caries is to limit the growth of cariogenic bacteria by using two anti-bacterial agents with different mechanisms of action. The hypothesis of this study was that the anti-bacterial activity of ω-6 polyunsaturated arachidonic acid (AA) against S. mutans and S. sobrinus can be enhanced by the sesquiterpene alcohol trans, trans-farnesol (t,t-farnesol). METHODS The anti-bacterial activity of single and combined treatment was determined by the checkerboard assay. Bacterial viability was assessed by live/dead SYTO 9/propidium iodide (PI) staining on flow cytometry. Anti-biofilm activity was determined by MTT metabolic assay, crystal violet staining of biofilm biomass, SYTO 9/PI staining by spinning disk confocal microscopy (SDCM) and high-resolution scanning electron microscopy (HR-SEM). RESULTS t,t-Farnesol lowered the minimum inhibitory concentration (MIC) and the minimum biofilm inhibitory concentration (MBIC) of AA at sub-MICs. AA reduced the metabolic activity of preformed mature biofilms, while t,t-farnesol had no significant effect. The enhanced anti-bacterial effect of the combined t,t-farnesol/AA treatment was further evidenced by increased PI uptake, indicating membrane perforation. The enhanced anti-biofilm effect was further verified by SDCM and HR-SEM. Gene expression studies showed reduced expression of some biofilm-related genes. CONCLUSIONS Altogether, our study suggests a potential use of the two naturally occurring compounds arachidonic acid and t,t-farnesol for preventing biofilm formation by the cariogenic bacteria S. mutans and S. sobrinus. These findings have implications for caries prevention.
Collapse
|
31
|
Yu R, Ai N, Huang C, Wang D, Bian C, Ge W, Chong CM. Aspirin reduces Ponatinib-induced cardiovascular toxic phenotypes and death in zebrafish. Biomed Pharmacother 2024; 180:117503. [PMID: 39357328 DOI: 10.1016/j.biopha.2024.117503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 09/20/2024] [Accepted: 09/25/2024] [Indexed: 10/04/2024] Open
Abstract
BACKGROUND Ponatinib (Iclusig) is an oral tyrosine kinase BCR-ABL inhibitor for treating patients with Philadelphia chromosome-positive acute lymphoblastic leukemia (Ph+ ALL) and chronic myeloid leukemia (CML) who are resistant to the therapies with other tyrosine kinase inhibitors. However, adverse cardiovascular events caused by Ponatinib are a serious issue that affects patients' survival rates. Thus, it is necessary to search for candidate drugs to reduce the cardiovascular toxicity of Ponatinib. PURPOSE To investigate the effects of Aspirin on Ponatinib-induced cardiovascular toxicity in zebrafish. METHODS AB strain of wild type zebrafish (Danio rerio), Tg (cmlc2: GFP) transgenic zebrafish, and Tg (gata1: dsRed) transgenic zebrafish were used as in vivo models to assess survival, blood flow, cardiac morphology, and function. Thrombus formation was detected using O-dianisidine staining. The transcriptome of zebrafish larvae treated with Ponatinib was assessed using RNA sequencing. RESULTS Ponatinib not only reduced survival rate but also caused cardiovascular toxic events such as pericardial edema, abnormal heart structure, low heart rate, and thrombosis. In addition, whole-body transcriptome analysis showed that Ponatinib up-regulated the expression of cyclooxygenase-1 (COX-1). Compared with other antithrombotic drugs, a COX-1 inhibitor Aspirin more effectively reduced ponatinib-induced cardiovascular toxicity events and improved the survival rate of zebrafish larvae. CONCLUSION Our findings suggest that Aspirin exhibits the potential to reduce Ponatinib-induced cardiovascular toxicity.
Collapse
Affiliation(s)
- Ruiqi Yu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau 999078, China
| | - Nana Ai
- Department of Biomedical Sciences and Centre of Reproduction, Development and Aging (CRDA), Faculty of Health Sciences, University of Macau, Taipa, Macau 999078, China
| | - Chen Huang
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, SAR 999078, China
| | - Danni Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau 999078, China
| | - Chao Bian
- Laboratory of Aquatic Genomics, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China
| | - Wei Ge
- Department of Biomedical Sciences and Centre of Reproduction, Development and Aging (CRDA), Faculty of Health Sciences, University of Macau, Taipa, Macau 999078, China.
| | - Cheong-Meng Chong
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau 999078, China.
| |
Collapse
|
32
|
Rajendran R, Krishnan R, Oh MJ. Viral reprogramming by nervous necrosis virus alters key metabolites and its pathways in sevenband grouper (Hyporthodus septemfasciatus) gills. FISH & SHELLFISH IMMUNOLOGY 2024; 154:109900. [PMID: 39265962 DOI: 10.1016/j.fsi.2024.109900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 09/03/2024] [Accepted: 09/09/2024] [Indexed: 09/14/2024]
Abstract
Nervous necrosis virus (NNV) which mainly infects sevenband grouper (Hyporthodus Septemfasciatus) is considered a potential threat to the grouper aquaculture industry. The gills being one of the portal of entry and an active site of replication of fish viruses emphasises its role as a key region to study the metabolomic changes caused by viral reprograming and hijacking of metabolic pathways associated with immunity of the host. In the present study, liquid chromatography mass spectrometry (LC-MS) was used to detect changes of endogenous compounds of the grouper after NNV infection. A total of 75 metabolites of ten different pathways were identified. The metabolites were mainly associated with fatty acids, lipids, amino acids and nucleotides. The virus reprogramming lead to the downregulation of majority of the metabolites in their pathways. Arachidonic acid (AA), tryptophan, kynurenine and methandriol were selected as representative metabolites and challenge studies with NNV confirmed the fact that, metabolites controlled the replication of virus in a dose dependent manner. Immune gene expression studies also confirmed the effect of metabolites by upregulated expression of interleukins, cytokines and TLRs which are part of cellular immune response. This study shows the viral reprogramming of NNV in grouper gill cells resulting in alterations in basic metabolic pathways associated with normal functioning of the organism.
Collapse
Affiliation(s)
- Rahul Rajendran
- Department of Aqualife Medicine, Chonnam National University, Yeosu, 50626, Republic of Korea
| | - Rahul Krishnan
- Department of Aquatic Animal Health Management, Kerala University of Fisheries and Ocean Studies, Kerala, 682506, India
| | - Myung-Joo Oh
- Department of Aqualife Medicine, Chonnam National University, Yeosu, 50626, Republic of Korea.
| |
Collapse
|
33
|
Tan D, Li E, Xiong S, Sun Y, Cheng W, Su Y, Lu Y. Transcriptomic and Metabolomic Analyses Reveal the Attenuating Role of Cordycepin and Cordyceps militaris Extract on Acute Liver Injury Induced by LPS in Piglets. Animals (Basel) 2024; 14:2873. [PMID: 39409822 PMCID: PMC11475243 DOI: 10.3390/ani14192873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 10/02/2024] [Accepted: 10/03/2024] [Indexed: 10/20/2024] Open
Abstract
Cordyceps militaris extract (CME) contains many bioactive compounds, mainly cordycepin (CPN). This study aimed to investigate the possible mechanisms underlying the amelioration of LPS-induced acute liver injury in piglets by CME or CPN supplementation using multi-omics analysis. Twenty-four weaned piglets were randomly distributed into 4 groups (n = 6): the control and LPS groups were fed basal diets; the CPN + LPS (CPN-LPS) and CME + LPS (CME-LPS) groups were fed the basal diets supplemented with CME or CPN. The results showed that CPN or CME supplementation significantly decreased the C-reactive protein level (p < 0.05) and improved liver tissue pathology to prevent acute liver injury after LPS treatment. Compared with LPS, the transcriptomic analysis indicated that CPN supplementation significantly downregulated cell adhesion molecules, while CME supplementation significantly downregulated inflammatory mediator regulation of TRP channels, complement and coagulation cascades and cytokine-cytokine receptor interaction. The metabolomic results showed that CPN or CME supplementation significantly reduced disease biomarker of bicyclo-prostaglandin E2, and increased levels of deoxyinosine and 3-hydroxyanthranilic acid (p < 0.05). The combined transcriptome and metabolome helped identify two metabolites PC 34:2 and PC 36:0, which may be associated with the restoration of liver cell morphology. In conclusion, CPN and CME could attenuate LPS-induced acute liver injury by regulating immune-related genes and metabolites. This study elucidates the potential protective mechanism of CPN or CME supplementation against acute liver injury.
Collapse
Affiliation(s)
- Ding Tan
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (D.T.); (E.L.); (S.X.); (W.C.)
| | - Endian Li
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (D.T.); (E.L.); (S.X.); (W.C.)
| | - Shijie Xiong
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (D.T.); (E.L.); (S.X.); (W.C.)
| | - Yue Sun
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (D.T.); (E.L.); (S.X.); (W.C.)
| | - Wenbo Cheng
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (D.T.); (E.L.); (S.X.); (W.C.)
| | - Yong Su
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (D.T.); (E.L.); (S.X.); (W.C.)
| | - Yang Lu
- Institute of Animal Husbandry and Veterinary Science, Shanghai Academy of Agricultural Sciences, Shanghai 201106, China
| |
Collapse
|
34
|
Qin X, Tan Z, Li Q, Zhang S, Hu D, Wang D, Wang L, Zhou B, Liao R, Wu Z, Liu Y. Rosiglitazone attenuates Acute Kidney Injury from hepatic ischemia-reperfusion in mice by inhibiting arachidonic acid metabolism through the PPAR-γ/NF-κB pathway. Inflamm Res 2024; 73:1765-1780. [PMID: 39112648 DOI: 10.1007/s00011-024-01929-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 07/30/2024] [Accepted: 08/02/2024] [Indexed: 10/02/2024] Open
Abstract
BACKGROUND Acute Kidney Injury (AKI), a prevalent complication of Liver Transplantation (LT) that occurs during the perioperative period has been established to profoundly impact the prognosis of transplant recipients. This study aimed to investigate the mechanism of the hepatic IRI-induced AKI and to identify potential therapeutic targets for treating this condition and improving the prognosis of LT patients. METHODS An integrated transcriptomics and proteomics approach was employed to investigate transcriptional and proteomic alterations in hepatic IRI-induced AKI and the hypoxia-reoxygenation (H/R) model using TCMK-1 cells and the hepatic IRI-induced AKI mouse model using male C57BL/6 J mice were employed to elucidate the underlying mechanisms. Hematoxylin-eosin staining, reverse transcription quantitative polymerase chain reaction, enzyme-linked immunosorbent assay and Western blot were used to assess the effect of Rosiglitazone (RGZ) on hepatic IRI-induced AKI in vitro and in vivo. RESULTS According to the results, 322 genes and 128 proteins were differentially expressed between the sham and AKI groups. Furthermore, Gene Ontology (GO) and the Kyoto Encyclopedia of Genes and Genomics (KEGG) pathway analyses revealed significant enrichment in pathways related to amino acid and lipid metabolism. Additionally, the Protein-Protein Interaction (PPI) network analysis of the kidney tissues obtained from a hepatic IRI-induced AKI mouse model highlighted arachidonic acid metabolism as the most prominent pathway. Animal and cellular analyses further revealed that RGZ, a PPAR-γ agonist, could inhibit the expression of the PPAR-γ/NF-κB signaling pathway-associated proteins in in vitro and in vivo. CONCLUSIONS These findings collectively suggest that RGZ ameliorates hepatic IRI-induced AKI via PPAR-γ/NF-κB signaling pathway modulation, highlighting PPAR-γ as a crucial therapeutic target for AKI prevention post-LT.
Collapse
Affiliation(s)
- Xiaoyan Qin
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400042, China
- Department of General Surgery and Trauma Surgery, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Chongqing, 400014, China
| | - Zhengli Tan
- The First Clinical College of Chongqing Medical University, Chongqing, 400046, China
| | - Qi Li
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400042, China
| | - Shiyi Zhang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400042, China
| | - Dingheng Hu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400042, China
| | - Denghui Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400042, China
| | - Liangxu Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400042, China
| | - Baoyong Zhou
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400042, China
| | - Rui Liao
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400042, China
| | - Zhongjun Wu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400042, China
| | - Yanyao Liu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400042, China.
| |
Collapse
|
35
|
Akram MZ, Sureda EA, Corion M, Comer L, Everaert N. Linking gastrointestinal tract structure, function, and gene expression signatures to growth variability in broilers: a novel interpretation for flock uniformity. Poult Sci 2024; 103:104158. [PMID: 39173569 PMCID: PMC11387703 DOI: 10.1016/j.psj.2024.104158] [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: 06/09/2024] [Revised: 07/29/2024] [Accepted: 07/29/2024] [Indexed: 08/24/2024] Open
Abstract
Variation in body weight (BW) within broiler flocks is a significant challenge in poultry production. Investigating differences in gut-related parameters between low (LBW) and high BW (HBW) chicks may provide insights into the underlying causes of BW heterogeneity. 908 day-old male broiler chicks were reared until d 7 and then ranked into LBW and HBW groups. Thereafter, performance parameters were compared between BW groups periodically. On d 7, 14, and 38, visceral organ characteristics, intestinal permeability, and duodenal and ileal histomorphology were examined. Expression profiles were analyzed for 79 ileal genes related to gut barrier function, immune function, nutrient transport, gut hormones, nutrient receptors, metabolism, and oxidation using high-throughput qPCR. Student's t-tests were performed to compare measurements. Multivariate statistics, including partial least square regression (PLSR) analysis, were applied to identify combinations of key genes discriminating BW groups, offering predictive capability for phenotypic variations. The HBW group remained heavier at each timepoint, which could be explained by higher feed intake. The HBW group had shorter relative small intestine length but higher villus height and villi height/crypt depth ratios. The LBW group demonstrated increased intestinal permeability on d 38. The LBW group showed upregulation of immune response genes including TNF-α on d 7 and CYP450 on d 38, while the HBW group showed higher AHSA1 and HSPA4 expressions on d 7. The LBW group had upregulation of the metabolism genes mTOR and EIF4EBP1 on d 7 and the satiety-induced hormone cholecystokinin on d 14, while the HBW group tended to increase expression of the hunger hormone ghrelin on d 38. Genes related to gut barrier function, nutrient transport, and oxidation categories were consistently upregulated in the HBW group. PLSR models revealed 4, 12, and 11 sets of key genes highly predictive of BW phenotypes on d 7, 14, and 38, respectively. These findings suggest that growth rates are linked to the intestinal size, structure, and function of broiler chickens, offering insights into the underlying mechanisms regulating BW.
Collapse
Affiliation(s)
- Muhammad Zeeshan Akram
- Nutrition and Animal-Microbiota Ecosystems Laboratory, Department of Biosystems, KU Leuven, 3000-Heverlee, Belgium; Precision Livestock and Nutrition Unit, Gembloux Agro-Bio Tech, University of Liège, Gembloux, Belgium
| | - Ester Arévalo Sureda
- Nutrition and Animal-Microbiota Ecosystems Laboratory, Department of Biosystems, KU Leuven, 3000-Heverlee, Belgium
| | - Matthias Corion
- Nutrition and Animal-Microbiota Ecosystems Laboratory, Department of Biosystems, KU Leuven, 3000-Heverlee, Belgium
| | - Luke Comer
- Nutrition and Animal-Microbiota Ecosystems Laboratory, Department of Biosystems, KU Leuven, 3000-Heverlee, Belgium
| | - Nadia Everaert
- Nutrition and Animal-Microbiota Ecosystems Laboratory, Department of Biosystems, KU Leuven, 3000-Heverlee, Belgium.
| |
Collapse
|
36
|
Ma Y, Zou C, Yang Y, Fang M, Guan Y, Sun J, Gao Y, Shang Z, Zhang X. Arachidonic acid enhances hepatocyte bile acid uptake and alleviates cholestatic liver disease by upregulating OATP1 expression. Food Funct 2024; 15:9916-9927. [PMID: 39258405 DOI: 10.1039/d4fo02158d] [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: 09/12/2024]
Abstract
Cholestatic liver disease is caused by disorders of bile synthesis, secretion, and excretion. Over the long term, progressive liver cell damage from the disease evolves into liver fibrosis and cirrhosis, ultimately leading to liver failure and even cancer. Notably, cholestatic liver disease has a complex pathogenesis that remains relatively unclear. In this study, we generated two mouse models of cholestatic liver disease using a 0.1% 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC) diet and α-naphthyl isothiocyanate (ANIT) gavage. Quantitative proteomics using liquid chromatography-tandem mass spectrometry showed that arachidonic acid metabolism was a common pathway in both models. Additionally, serum arachidonic acid concentrations were lower in both models than in the control group. Arachidonic acid supplementation in the diet of DDC model mice significantly reduced the levels of serum markers of cholestasis (alanine aminotransferase, aspartate transaminase, alkaline phosphatase, total bile acid, and total bilirubin) and decreased the degree of bile duct hyperplasia and cholestasis. To elucidate the mechanisms by which arachidonic acid improved bile stasis, we analyzed gene expression after arachidonic acid administration and found that Oatp1 was upregulated in the liver tissue of cholestatic mice. Arachidonic acid also increased Oatp1 expression in AML12 cells, which promoted bile acid uptake. Conclusively, our research showed that arachidonic acid mitigates cholestatic liver disease by upregulating Oatp1, promoting bile acid uptake by hepatocytes and participating in intestinal-hepatic circulation. Overall, these results suggest that supplementing foods with arachidonic acid in the daily diet may be an effective treatment strategy for cholestatic liver disease.
Collapse
Affiliation(s)
- Yanlu Ma
- Laboratory of Cellular Immunity, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
- Shanghai Traditional Chinese Medicine Clinical Key Laboratory, Shanghai, China
| | - Chen Zou
- Laboratory of Cellular Immunity, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
- Shanghai Traditional Chinese Medicine Clinical Key Laboratory, Shanghai, China
| | - Yilan Yang
- Laboratory of Cellular Immunity, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
- Shanghai Traditional Chinese Medicine Clinical Key Laboratory, Shanghai, China
| | - Miao Fang
- Laboratory of Cellular Immunity, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
- Shanghai Traditional Chinese Medicine Clinical Key Laboratory, Shanghai, China
| | - Yunfeng Guan
- Laboratory of Cellular Immunity, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
- Shanghai Traditional Chinese Medicine Clinical Key Laboratory, Shanghai, China
| | - Jianqi Sun
- Laboratory of Cellular Immunity, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
- Shanghai Traditional Chinese Medicine Clinical Key Laboratory, Shanghai, China
| | - Yueqiu Gao
- Laboratory of Cellular Immunity, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
- Shanghai Traditional Chinese Medicine Clinical Key Laboratory, Shanghai, China
- Department of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine Anhui Hospital, Anhui, China
| | - Zhi Shang
- Laboratory of Cellular Immunity, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
- Shanghai Traditional Chinese Medicine Clinical Key Laboratory, Shanghai, China
| | - Xin Zhang
- Laboratory of Cellular Immunity, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
- Shanghai Traditional Chinese Medicine Clinical Key Laboratory, Shanghai, China
- Department of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
37
|
Josyula JVN, JeanPierre AR, Jorvekar SB, Adla D, Mariappan V, Pulimamidi SS, Green SR, Pillai AB, Borkar RM, Mutheneni SR. Metabolomic profiling of dengue infection: unraveling molecular signatures by LC-MS/MS and machine learning models. Metabolomics 2024; 20:104. [PMID: 39305446 DOI: 10.1007/s11306-024-02169-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Accepted: 09/02/2024] [Indexed: 10/22/2024]
Abstract
BACKGROUND & OBJECTIVE The progression of dengue fever to severe dengue (SD) is a major public health concern that impairs the capacity of the medical system to predict and treat dengue patients. Hence, the present study used a metabolomic approach integrated with machine models to identify differentially expressed metabolites in patients with SD compared to nonsevere patients and healthy controls. METHODS Comprehensively, the plasma was collected at different clinical phases during dengue without warning signs (DWOW, N = 10), dengue with warning signs (DWW, N = 10), and SD (N = 10) at different stages [i.e., day of admission (DOA), day of defervescence (DOD), and day of convalescent (DOC)] in comparison to healthy control (HC). The samples were subjected to LC‒ESI‒MS/MS to identify metabolites. Statistical and machine learning analyses were performed using R and Python language. Further, biomarker, pathway and correlation analysis was performed to identify potential predictors of dengue. RESULTS & CONCLUSION A total of 423 metabolites were identified in all the study groups. Paired and unpaired t-tests revealed 14 highly differentially expressed metabolites between and across the dengue groups, with four metabolites (shikimic acid, ureidosuccinic acid, propionyl carnitine, and alpha-tocopherol) showing significant differences compared to HC. Furthermore, biomarker (ROC) analysis revealed 11 potential molecules with a significant AUC value of 1 that could serve as potential biomarkers for identifying different dengue clinical stages that are beneficial for predicting dengue disease outcomes. The logistic regression model revealed that S-adenosylhomocysteine, hypotaurine, and shikimic acid metabolites could be beneficial indicators for predicting severe dengue, with an accuracy and AUC of 0.75. The data showed that dengue infection is related to lipid metabolism, oxidative stress, inflammation, metabolomic adaptation, and virus manipulation. Moreover, the biomarkers had a significant correlation with biochemical parameters like platelet count, and hematocrit. These results shed some light on host-derived small-molecule biomarkers that are associated with dengue severity and novel insights into metabolomics mechanisms interlinked with disease severity.
Collapse
Affiliation(s)
- Jhansi Venkata Nagamani Josyula
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Tarnaka, Hyderabad, Telangana, 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Aashika Raagavi JeanPierre
- Mahatma Gandhi Medical Advanced Research Institute (MGMARI), Sri Balaji Vidyapeeth (Deemed to be University), Puducherry, 607 402, India
| | - Sachin B Jorvekar
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research, Guwahati, Assam, 781101, India
| | - Deepthi Adla
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Tarnaka, Hyderabad, Telangana, 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Vignesh Mariappan
- Mahatma Gandhi Medical Advanced Research Institute (MGMARI), Sri Balaji Vidyapeeth (Deemed to be University), Puducherry, 607 402, India
| | - Sai Sharanya Pulimamidi
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research, Guwahati, Assam, 781101, India
| | - Siva Ranganathan Green
- Mahatma Gandhi Medical College and Research Institute (MGMCRI), Sri Balaji Vidyapeeth (Deemed to be University), Puducherry, 607 402, India
| | - Agieshkumar Balakrishna Pillai
- Mahatma Gandhi Medical Advanced Research Institute (MGMARI), Sri Balaji Vidyapeeth (Deemed to be University), Puducherry, 607 402, India.
| | - Roshan M Borkar
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research, Guwahati, Assam, 781101, India
| | - Srinivasa Rao Mutheneni
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Tarnaka, Hyderabad, Telangana, 500007, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
38
|
Alfadda AA, Abdel Rahman AM, Benabdelkamel H, AlMalki R, Alsuwayni B, Alhossan A, Aldhwayan MM, Abdeen GN, Miras AD, Masood A. Metabolomic Effects of Liraglutide Therapy on the Plasma Metabolomic Profile of Patients with Obesity. Metabolites 2024; 14:500. [PMID: 39330507 PMCID: PMC11433991 DOI: 10.3390/metabo14090500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 09/09/2024] [Accepted: 09/14/2024] [Indexed: 09/28/2024] Open
Abstract
BACKGROUND Liraglutide, a long-acting glucagon-like peptide-1 receptor agonist (GLP1RA), is a well-established anti-diabetic drug, has also been approved for the treatment of obesity at a dose of 3 mg. There are a limited number of studies in the literature that have looked at changes in metabolite levels before and after liraglutide treatment in patients with obesity. To this end, in the present study we aimed to explore the changes in the plasma metabolomic profile, using liquid chromatography-high resolution mass spectrometry (LC-HRMS) in patients with obesity. METHODS A single-center prospective study was undertaken to evaluate the effectiveness of 3 mg liraglutide therapy in twenty-three patients (M/F: 8/15) with obesity, mean BMI 40.81 ± 5.04 kg/m2, and mean age of 36 ± 10.9 years, in two groups: at baseline (pre-treatment) and after 12 weeks of treatment (post-treatment). An untargeted metabolomic profiling was conducted in plasma from the pre-treatment and post-treatment groups using LC-HRMS, along with bioinformatics analysis using ingenuity pathway analysis (IPA). RESULTS The metabolomics analysis revealed a significant (FDR p-value ≤ 0.05, FC 1.5) dysregulation of 161 endogenous metabolites (97 upregulated and 64 downregulated) with distinct separation between the two groups. Among the significantly dysregulated metabolites, the majority of them were identified as belonging to the class of oxidized lipids (oxylipins) that includes arachidonic acid and its derivatives, phosphorglycerophosphates, N-acylated amino acids, steroid hormones, and bile acids. The biomarker analysis conducted using MetaboAnalyst showed PGP (a21:0/PG/F1alpha), an oxidized lipid, as the first metabolite among the list of the top 15 biomarkers, followed by cysteine and estrone. The IPA analysis showed that the dysregulated metabolites impacted the pathway related to cell signaling, free radical scavenging, and molecular transport, and were focused around the dysregulation of NF-κB, ERK, MAPK, PKc, VEGF, insulin, and pro-inflammatory cytokine signaling pathways. CONCLUSIONS The findings suggest that liraglutide treatment reduces inflammation and modulates lipid metabolism and oxidative stress. Our study contributes to a better understanding of the drug's multifaceted impact on overall metabolism in patients with obesity.
Collapse
Affiliation(s)
- Assim A. Alfadda
- Proteomics Resource Unit, Obesity Research Center, College of Medicine, King Saud University, Riyadh 11461, Saudi Arabia; (H.B.); (A.M.)
- Strategic Center for Diabetes Research, College of Medicine, King Saud University, Riyadh 11461, Saudi Arabia
- Department of Medicine, College of Medicine, King Saud University, Riyadh 11461, Saudi Arabia
| | - Anas M. Abdel Rahman
- Metabolomics Section, Department of Clinical Genomics, Center for Genomics Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia; (A.M.A.R.); (R.A.)
| | - Hicham Benabdelkamel
- Proteomics Resource Unit, Obesity Research Center, College of Medicine, King Saud University, Riyadh 11461, Saudi Arabia; (H.B.); (A.M.)
| | - Reem AlMalki
- Metabolomics Section, Department of Clinical Genomics, Center for Genomics Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia; (A.M.A.R.); (R.A.)
| | - Bashayr Alsuwayni
- Corporate of Pharmacy Services, King Saud University Medical City, Riyadh 11461, Saudi Arabia;
| | - Abdulaziz Alhossan
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh 11461, Saudi Arabia;
| | - Madhawi M. Aldhwayan
- Department of Community Health Science, Clinical Nutrition, College of Applied Medical Sciences, King Saud University, Riyadh 11461, Saudi Arabia; (M.M.A.); (G.N.A.)
| | - Ghalia N. Abdeen
- Department of Community Health Science, Clinical Nutrition, College of Applied Medical Sciences, King Saud University, Riyadh 11461, Saudi Arabia; (M.M.A.); (G.N.A.)
| | - Alexander Dimitri Miras
- Section of Investigative Medicine, Division of Diabetes, Endocrinology and Metabolic Medicine, Hammersmith Hospital, Imperial College London, London SW7 2AZ, UK;
- School of Medicine, Ulster University, Derry BT1 6DN, UK
| | - Afshan Masood
- Proteomics Resource Unit, Obesity Research Center, College of Medicine, King Saud University, Riyadh 11461, Saudi Arabia; (H.B.); (A.M.)
| |
Collapse
|
39
|
Siavoshi F, Ladakis DC, Muller A, Nourbakhsh B, Bhargava P. Ocrelizumab alters the circulating metabolome in people with relapsing-remitting multiple sclerosis. Ann Clin Transl Neurol 2024; 11:2485-2498. [PMID: 39185939 PMCID: PMC11537130 DOI: 10.1002/acn3.52167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 07/09/2024] [Accepted: 07/12/2024] [Indexed: 08/27/2024] Open
Abstract
BACKGROUND Circulating metabolite levels are altered in multiple sclerosis (MS) and are associated with MS severity. However, how metabolic profiles shift following highly efficacious therapies, like ocrelizumab remains unclear. OBJECTIVE Circulating metabolite levels are altered in multiple sclerosis (MS) and are associated with MS severity. However, how metabolic profiles shift following highly efficacious therapies, like ocrelizumab remains unclear. To assess changes in the circulating metabolome produced by ocrelizumab treatment in people with relapsing-remitting MS (RRMS). METHODS Thirty-one individuals with RRMS eligible for beginning treatment with ocrelizumab were recruited and followed with demographic, clinical, quality-of-life, and global metabolomics data collected at each visit. Modules of highly correlated metabolites were identified using the weighted correlation network analysis approach. Changes in each module's eigenmetabolite values and individual metabolites during the study were evaluated using linear mixed-effects models. RESULTS Patients with a mean age of 40.8 (SD = 10.30) years, and median disease duration of 4.0 (IQR = 8.5) years, were monitored for a median of 3.36 (IQR = 1.43) years. Two out of twelve identified sets of metabolites were altered significantly. The first module mainly contained androgenic and pregnenolone steroids (p-value <0.001, coefficient: -0.10). The second module primarily consisted of several lysophospholipids, arachidonic acid, some endocannabinoids, and monohydroxy fatty acid metabolites (p-value = 0.016, coefficient: -0.12), which its reduction was significantly associated with improvement based on overall disability response score (OR 3.09e-01, 95% CI: 6.83e-02, 9.09e-01, p-value = 3.15E-02). INTERPRETATION In this longitudinal observational study, using a global untargeted metabolomics approach, we showed significant alteration in circulating metabolome in RRMS patients undergoing ocrelizumab treatment. In particular, we observed a significant reduction in metabolites involved in the lysophospholipid pathway, which was associated with patients' improvement.
Collapse
Affiliation(s)
- Fatemeh Siavoshi
- Department of NeurologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Dimitrios C. Ladakis
- Department of NeurologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Ashley Muller
- Department of NeurologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Bardia Nourbakhsh
- Department of NeurologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Pavan Bhargava
- Department of NeurologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| |
Collapse
|
40
|
Rivi V, Rigillo G, Batabyal A, Lukowiak K, Pani L, Tascedda F, Benatti C, Blom JMC. Different stressors uniquely affect the expression of endocannabinoid-metabolizing enzymes in the central ring ganglia of Lymnaea stagnalis. J Neurochem 2024; 168:2848-2867. [PMID: 38922726 DOI: 10.1111/jnc.16147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 04/16/2024] [Accepted: 05/16/2024] [Indexed: 06/28/2024]
Abstract
The endocannabinoid system (ECS) plays an important role in neuroprotection, neuroplasticity, energy balance, modulation of stress, and inflammatory responses, acting as a critical link between the brain and the body's peripheral regions, while also offering promising potential for novel therapeutic strategies. Unfortunately, in humans, pharmacological inhibitors of different ECS enzymes have led to mixed results in both preclinical and clinical studies. As the ECS has been highly conserved throughout the eukaryotic lineage, the use of invertebrate model organisms like the pond snail Lymnaea stagnalis may provide a flexible tool to unravel unexplored functions of the ECS at the cellular, synaptic, and behavioral levels. In this study, starting from the available genome and transcriptome of L. stagnalis, we first identified putative transcripts of all ECS enzymes containing an open reading frame. Each predicted protein possessed a high degree of sequence conservation to known orthologues of other invertebrate and vertebrate organisms. Sequences were confirmed by qualitative PCR and sequencing. Then, we investigated the transcriptional effects induced by different stress conditions (i.e., bacterial LPS injection, predator scent, food deprivation, and acute heat shock) on the expression levels of the enzymes of the ECS in Lymnaea's central ring ganglia. Our results suggest that in Lymnaea as in rodents, the ECS is involved in mediating inflammatory and anxiety-like responses, promoting energy balance, and responding to acute stressors. To our knowledge, this study offers the most comprehensive analysis so far of the ECS in an invertebrate model organism.
Collapse
Affiliation(s)
- Veronica Rivi
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Giovanna Rigillo
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Anuradha Batabyal
- Department of Physical and Natural Sciences, FLAME University, Pune, India
| | - Ken Lukowiak
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Luca Pani
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
- Department of Psychiatry and Behavioral Sciences, University of Miami, Miami, Florida, USA
| | - Fabio Tascedda
- Centre of Neuroscience and Neurotechnology, University of Modena and Reggio Emilia, Modena, Italy
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
- CIB, Consorzio Interuniversitario Biotecnologie, Trieste, Italy
| | - Cristina Benatti
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
- Centre of Neuroscience and Neurotechnology, University of Modena and Reggio Emilia, Modena, Italy
| | - Johanna M C Blom
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
- Centre of Neuroscience and Neurotechnology, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
41
|
Liu Q, Wu X, Liu C, Wang N, Yin F, Wu H, Cao S, Zhao W, Wu H, Zhou A. Metabolomic and biochemical changes in the plasma and liver of toxic milk mice model of Wilson disease. J Pharm Biomed Anal 2024; 246:116255. [PMID: 38795427 DOI: 10.1016/j.jpba.2024.116255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 05/11/2024] [Accepted: 05/23/2024] [Indexed: 05/28/2024]
Abstract
Wilson disease (WD) is an inherited disorder characterized by abnormal copper metabolism with complex pathological features. Currently, this mechanism of copper overload-induced hepatic injury remains unclear. In this study, male toxic milk (TX) mice were selected as experimental subjects. Copper levels and biochemical indices were measured by atomic absorption spectroscopy (AAS) and kits. Liver tissue ultrastructure was observed by hematoxylin-eosin (H&E), sirius red staining and transmission electron microscopy. Plasma and liver metabolic profiles of TX mice were characterized by untargeted metabolomics. In addition, the expression of enzymes related to arachidonic acid metabolism in liver tissue was detected by Western blotting. The results showed the excessive copper content, concomitant oxidative stress, and hepatic tissue structural damage in TX mice. Seventy-eight metabolites were significantly different in WD, mainly involved in the metabolism of arachidonic acid, glycerophospholipids, sphingolipids, niacin and nicotinamide, and phenylalanine. Furthermore, the arachidonic acid metabolic pathway is an important pathway involved in WD metabolism. The level of arachidonic acid in the liver of TX mice was significantly lower (p < 0.01) compared to the control group. The expression of cytoplasmic phospholipase A2 (cPLA2) and arachidonic acid 12-lipoxygenase (ALOX12), related to the arachidonic acid metabolic pathway, was significantly different in the liver of TX mice (p < 0.01). Modulation of the arachidonic acid metabolic pathway could be a potential therapeutic strategy to alleviate WD symptoms.
Collapse
Affiliation(s)
- Qiao Liu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230038, China
| | - Xiaoyuan Wu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230038, China
| | - Cuicui Liu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230038, China
| | - Ni Wang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230038, China
| | - Fengxia Yin
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230038, China
| | - Huan Wu
- The Experimental Research Center, Anhui University of Chinese Medicine, Hefei 230038, China; Anhui Province Key Laboratory of Research and Development of Chinese Medicine, Functional Activity and Resource Utilization on Edible and Medicinal Fungi Joint Laboratory of Anhui Province, Hefei 230038, China
| | - Shijian Cao
- The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230031, China
| | - Wenchen Zhao
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh 15219, USA
| | - Hongfei Wu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230038, China; Anhui Province Key Laboratory of Research and Development of Chinese Medicine, Functional Activity and Resource Utilization on Edible and Medicinal Fungi Joint Laboratory of Anhui Province, Hefei 230038, China.
| | - An Zhou
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230038, China; Anhui Province Key Laboratory of Research and Development of Chinese Medicine, Functional Activity and Resource Utilization on Edible and Medicinal Fungi Joint Laboratory of Anhui Province, Hefei 230038, China.
| |
Collapse
|
42
|
Zhou Y, Gong J, Deng X, Shen L, Liu L. Novel insights: crosstalk with non-puerperal mastitis and immunity. Front Immunol 2024; 15:1431681. [PMID: 39148739 PMCID: PMC11324573 DOI: 10.3389/fimmu.2024.1431681] [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: 05/12/2024] [Accepted: 07/16/2024] [Indexed: 08/17/2024] Open
Abstract
The two primary types of non-puerperal mastitis (NPM) are granulomatous lobular mastitis (GLM) and plasma cell mastitis (PCM). Existing research indicates that immune inflammatory response is considered to be the core of the pathogenesis of GLM and PCM, and both innate and adaptive immune responses play an important role in the pathophysiology of PCM and GLM. However, the regulatory balance between various immune cells in these diseases is still unclear. Consequently, we present a comprehensive summary of the immune-related variables and recent advances in GLM and PCM.
Collapse
Affiliation(s)
- Yao Zhou
- Department of Galactophore, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
- Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Jie Gong
- Department of Galactophore, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
- Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Xianguang Deng
- Department of Galactophore, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
- Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Lele Shen
- Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Lifang Liu
- Department of Galactophore, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| |
Collapse
|
43
|
Karagiannis TC, Ververis K, Liang JJ, Pitsillou E, Kagarakis EA, Yi DTZ, Xu V, Hung A, El-Osta A. Investigation of the Anti-Inflammatory Properties of Bioactive Compounds from Olea europaea: In Silico Evaluation of Cyclooxygenase Enzyme Inhibition and Pharmacokinetic Profiling. Molecules 2024; 29:3502. [PMID: 39124908 PMCID: PMC11314539 DOI: 10.3390/molecules29153502] [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: 06/11/2024] [Revised: 07/22/2024] [Accepted: 07/24/2024] [Indexed: 08/12/2024] Open
Abstract
In a landmark study, oleocanthal (OLC), a major phenolic in extra virgin olive oil (EVOO), was found to possess anti-inflammatory activity similar to ibuprofen, involving inhibition of cyclooxygenase (COX) enzymes. EVOO is a rich source of bioactive compounds including fatty acids and phenolics; however, the biological activities of only a small subset of compounds associated with Olea europaea have been explored. Here, the OliveNetTM library (consisting of over 600 compounds) was utilized to investigate olive-derived compounds as potential modulators of the arachidonic acid pathway. Our first aim was to perform enzymatic assays to evaluate the inhibitory activity of a selection of phenolic compounds and fatty acids against COX isoforms (COX-1 and COX-2) and 15-lipoxygenase (15-LOX). Olive compounds were found to inhibit COX isoforms, with minimal activity against 15-LOX. Subsequent molecular docking indicated that the olive compounds possess strong binding affinities for the active site of COX isoforms, and molecular dynamics (MD) simulations confirmed the stability of binding. Moreover, olive compounds were predicted to have favorable pharmacokinetic properties, including a readiness to cross biological membranes as highlighted by steered MD simulations and umbrella sampling. Importantly, olive compounds including OLC were identified as non-inhibitors of the human ether-à-go-go-related gene (hERG) channel based on patch clamp assays. Overall, this study extends our understanding of the bioactivity of Olea-europaea-derived compounds, many of which are now known to be, at least in part, accountable for the beneficial health effects of the Mediterranean diet.
Collapse
Affiliation(s)
- Tom C. Karagiannis
- Epigenetics in Human Health and Disease Program, Baker Heart and Diabetes Institute, 75 Commercial Road, Prahran, VIC 3004, Australia
- Epigenomic Medicine Laboratory at prospED Polytechnic, Carlton, VIC 3053, Australia
- Department of Clinical Pathology, The University of Melbourne, Parkville, VIC 3010, Australia
- Department of Microbiology and Immunology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Katherine Ververis
- Epigenomic Medicine Laboratory at prospED Polytechnic, Carlton, VIC 3053, Australia
- Department of Clinical Pathology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Julia J. Liang
- Epigenetics in Human Health and Disease Program, Baker Heart and Diabetes Institute, 75 Commercial Road, Prahran, VIC 3004, Australia
- Epigenomic Medicine Laboratory at prospED Polytechnic, Carlton, VIC 3053, Australia
- School of Science, STEM College, RMIT University, Melbourne, VIC 3001, Australia
| | - Eleni Pitsillou
- Epigenomic Medicine Laboratory at prospED Polytechnic, Carlton, VIC 3053, Australia
- School of Science, STEM College, RMIT University, Melbourne, VIC 3001, Australia
| | - Evan A. Kagarakis
- Epigenomic Medicine Laboratory at prospED Polytechnic, Carlton, VIC 3053, Australia
| | - Debbie T. Z. Yi
- Epigenomic Medicine Laboratory at prospED Polytechnic, Carlton, VIC 3053, Australia
| | - Vivian Xu
- Epigenomic Medicine Laboratory at prospED Polytechnic, Carlton, VIC 3053, Australia
| | - Andrew Hung
- School of Science, STEM College, RMIT University, Melbourne, VIC 3001, Australia
| | - Assam El-Osta
- Epigenetics in Human Health and Disease Program, Baker Heart and Diabetes Institute, 75 Commercial Road, Prahran, VIC 3004, Australia
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Sha Tin, Hong Kong SAR, China
- Hong Kong Institute of Diabetes and Obesity, Prince of Wales Hospital, The Chinese University of Hong Kong, 3/F Lui Che Woo Clinical Sciences Building, 30–32 Ngan Shing Street, Sha Tin, Hong Kong SAR, China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Sha Tin, Hong Kong SAR, China
- Biomedical Laboratory Science, Department of Technology, Faculty of Health, University College Copenhagen, 2200 Copenhagen, Denmark
| |
Collapse
|
44
|
Aradhyula V, Breidenbach JD, Khatib-Shahidi BZ, Slogar JN, Eyong SA, Faleel D, Dube P, Gupta R, Khouri SJ, Haller ST, Kennedy DJ. Transcriptomic Analysis of Arachidonic Acid Pathway Genes Provides Mechanistic Insight into Multi-Organ Inflammatory and Vascular Diseases. Genes (Basel) 2024; 15:954. [PMID: 39062733 PMCID: PMC11275336 DOI: 10.3390/genes15070954] [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: 06/22/2024] [Revised: 07/15/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
Arachidonic acid (AA) metabolites have been associated with several diseases across various organ systems, including the cardiovascular, pulmonary, and renal systems. Lipid mediators generated from AA oxidation have been studied to control macrophages, T-cells, cytokines, and fibroblasts, and regulate inflammatory mediators that induce vascular remodeling and dysfunction. AA is metabolized by cyclooxygenase (COX), lipoxygenase (LOX), and cytochrome P450 (CYP) to generate anti-inflammatory, pro-inflammatory, and pro-resolutory oxidized lipids. As comorbid states such as diabetes, hypertension, and obesity become more prevalent in cardiovascular disease, studying the expression of AA pathway genes and their association with these diseases can provide unique pathophysiological insights. In addition, the AA pathway of oxidized lipids exhibits diverse functions across different organ systems, where a lipid can be both anti-inflammatory and pro-inflammatory depending on the location of metabolic activity. Therefore, we aimed to characterize the gene expression of these lipid enzymes and receptors throughout multi-organ diseases via a transcriptomic meta-analysis using the Gene Expression Omnibus (GEO) Database. In our study, we found that distinct AA pathways were expressed in various comorbid conditions, especially those with prominent inflammatory risk factors. Comorbidities, such as hypertension, diabetes, and obesity appeared to contribute to elevated expression of pro-inflammatory lipid mediator genes. Our results demonstrate that expression of inflammatory AA pathway genes may potentiate and attenuate disease; therefore, we suggest further exploration of these pathways as therapeutic targets to improve outcomes.
Collapse
Affiliation(s)
- Vaishnavi Aradhyula
- Department of Medicine, The University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Joshua D. Breidenbach
- Department of Medicine, The University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
- Biochemistry and Biotechnology Group, Bioscience Division, Los Alamos National Laboratory, Los Alamos, NM 87545, USA
| | - Bella Z. Khatib-Shahidi
- Department of Medicine, The University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Julia N. Slogar
- Department of Medicine, The University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Sonia A. Eyong
- Department of Medicine, The University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Dhilhani Faleel
- Department of Medicine, The University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Prabhatchandra Dube
- Department of Medicine, The University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Rajesh Gupta
- Department of Medicine, The University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Samer J. Khouri
- Department of Medicine, The University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Steven T. Haller
- Department of Medicine, The University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - David J. Kennedy
- Department of Medicine, The University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| |
Collapse
|
45
|
Ezechukwu HC, Ney LJ, Jarvis MA, Shrestha N, Holland OJ, Cuffe JSM, Perkins AV, Yau SY, McAinch AJ, Hryciw DH. Sex-Specific Changes to Brain Fatty Acids, Plasmalogen, and Plasma Endocannabinoids in Offspring Exposed to Maternal and Postnatal High-Linoleic-Acid Diets. Int J Mol Sci 2024; 25:7911. [PMID: 39063152 PMCID: PMC11277558 DOI: 10.3390/ijms25147911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 07/06/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
Linoleic acid (LA) is required for neuronal development. We have previously demonstrated sex-specific changes in cardiovascular and hepatic function in rat offspring from mothers consuming a high-LA diet, with some effects associated with reduced LA concentration in the postnatal diet. At this time, the impact of a high-maternal-LA diet on offspring brain development and the potential for the postnatal diet to alter any adverse changes are unknown. Rat offspring from mothers fed low- (LLA) or high-LA (HLA) diets during pregnancy and lactation were weaned at postnatal day 25 (PN25) and fed LLA or HLA diets until sacrifice in adulthood (PN180). In the offspring's brains, the postnatal HLA diet increased docosapentaenoate in males. The maternal HLA diet increased LA, arachidonate, docosapentaenoate, C18:0 dimethylacetal (DMA), C16:0 DMA, C16:0 DMA/C16:0, and C18:0 DMA/C18:0, but decreased eoicosenoate, nervoniate, lignocerate, and oleate in males. Maternal and postnatal HLA diets reduced oleate and vaccenate and had an interaction effect on myristate, palmitoleate, and eicosapentaenoate in males. In females, maternal HLA diet increased eicosadienoate. Postnatal HLA diet increased stearate and docosapentaenoate. Maternal and postnatal HLA diets had an interaction effect on oleate, arachidate, and docosahexaenoic acid (DHA)/omega (n)-6 docosapentaenoic acid (DPA) in females. Postnatal HLA diet decreased DHA/n-6 DPA in males and females. Postnatal HLA diet increased plasma endocannabinoids (arachidonoyl ethanolamide and 2-arachidonoyl glycerol), as well as other N-acyl ethanolamides and testosterone. HLA diet alters brain fatty acids, plasma endocannabinoids, and plasmalogen concentrations in a development-specific and sex-specific manner.
Collapse
Affiliation(s)
- Henry C. Ezechukwu
- School of Human Sciences, The University of Western Australia, Perth, WA 6009, Australia;
| | - Luke J. Ney
- School of Psychology and Counselling, Queensland University of Technology, Kelvin Grove, QLD 4059, Australia; (L.J.N.); (M.A.J.)
| | - Madeline A. Jarvis
- School of Psychology and Counselling, Queensland University of Technology, Kelvin Grove, QLD 4059, Australia; (L.J.N.); (M.A.J.)
| | - Nirajan Shrestha
- School of Pharmacy and Medical Science, Griffith University, Gold Coast, QLD 4222, Australia; (N.S.); (O.J.H.); (A.V.P.)
| | - Olivia J. Holland
- School of Pharmacy and Medical Science, Griffith University, Gold Coast, QLD 4222, Australia; (N.S.); (O.J.H.); (A.V.P.)
| | - James S. M. Cuffe
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia;
| | - Anthony V. Perkins
- School of Pharmacy and Medical Science, Griffith University, Gold Coast, QLD 4222, Australia; (N.S.); (O.J.H.); (A.V.P.)
- School of Health, University of Sunshine Coast, Sippy Downs, QLD 4556, Australia
| | - Suk-Yu Yau
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Kowloon, Hong Kong;
- Mental Health Research Center, The Hong Kong Polytechnic University, Kowloon, Hong Kong
| | - Andrew J. McAinch
- Institute for Health and Sport, Victoria University, Melbourne, VIC 8001, Australia;
- Australian Institute for Musculoskeletal Science (AIMSS), Victoria University, St. Albans, VIC 3021, Australia
| | - Deanne H. Hryciw
- School of Environment and Science, Griffith University, Nathan, QLD 4111, Australia
- Griffith Institute for Drug Discovery, Griffith University, Nathan, QLD 4111, Australia
| |
Collapse
|
46
|
Xue Y, Lu J, Liu Y, Gao Y, Gong Y, Yang Y, Xiong Y, Shi X. Dihydroartemisinin modulated arachidonic acid metabolism and mitigated liver inflammation by inhibiting the activation of 5-LOX and COX-2. Heliyon 2024; 10:e33370. [PMID: 39027511 PMCID: PMC11255665 DOI: 10.1016/j.heliyon.2024.e33370] [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: 03/19/2024] [Revised: 06/19/2024] [Accepted: 06/20/2024] [Indexed: 07/20/2024] Open
Abstract
Background Dihydroartemisinin (DHA), a derivative of Artemisia annua, has been shown to possess anti-inflammatory properties. Besides, Yes-associated protein 1 (YAP1) plays a crucial role in maintaining liver homeostasis. Methods This study used Yap1 Flox/Flox, Albumin-Cre mice with hepatocyte-specific Yap1 knockout (referred to as Yap1 LKO) and their control mice (Yap1 Flox/Flox, referred to as Yap1 Flox). The effect of Yap1 on lipid metabolism homeostasis was investigated through non-targeted metabolomic analysis of mouse liver. Subsequently, DHA was administered to Yap1 LKO mice to assess its potential as a treatment. Liver pathology was evaluated via H&E staining, and the levels of AST, ALT, and TG were quantified using biochemical assays. The contents of arachidonic acid (AA), prostaglandin E1 (PGE1), and leukotrienes (LT) in the liver were measured using ELISA, while the protein expressions of PLIN2, 5-lipoxygenase (5-LOX), and cyclooxygenase-2 (COX-2) were analyzed through IHC staining. Results Hepatocyte-specific Yap1 knockout activated the AA metabolic pathway, resulting in increased elevated levels of AA, PGE1, and LT levels, along with inflammatory cytokine infiltration. DHA mitigated the elevation of metabolites such as PGE1 and LT caused by the AA metabolic pathway activation by down-regulating the levels of COX-2 and 5-LOX in the liver of Yap1 LKO mice. Moreover, it alleviated the accumulation of lipid vacuoles and reduced triglyceride (TG) and perilipin-2 (PLIN2) levels in the liver of Yap1 LKO mice. Conclusions Excessively low YAP1 expression induces liver inflammation and disturbances in lipid metabolism, whereas DHA modulated AA metabolism and mitigated liver inflammation by inhibiting the activation of 5-LOX and COX-2.
Collapse
Affiliation(s)
- Yu Xue
- Laboratory of Integrated Medicine Tumor Immunology, Shanxi University of Chinese Medicine, Taiyuan, 030000, China
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang, 050200, China
| | - Junlan Lu
- Laboratory of Integrated Medicine Tumor Immunology, Shanxi University of Chinese Medicine, Taiyuan, 030000, China
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang, 050200, China
| | - Yiwei Liu
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang, 050200, China
| | - Yuting Gao
- Laboratory of Integrated Medicine Tumor Immunology, Shanxi University of Chinese Medicine, Taiyuan, 030000, China
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang, 050200, China
| | - Yi Gong
- Laboratory of Integrated Medicine Tumor Immunology, Shanxi University of Chinese Medicine, Taiyuan, 030000, China
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang, 050200, China
| | - Yanguang Yang
- Laboratory of Integrated Medicine Tumor Immunology, Shanxi University of Chinese Medicine, Taiyuan, 030000, China
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang, 050200, China
| | - Yajun Xiong
- Laboratory of Integrated Medicine Tumor Immunology, Shanxi University of Chinese Medicine, Taiyuan, 030000, China
| | - Xinli Shi
- Laboratory of Integrated Medicine Tumor Immunology, Shanxi University of Chinese Medicine, Taiyuan, 030000, China
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang, 050200, China
| |
Collapse
|
47
|
Chang H, Zhang X, Lu Z, Gao B, Shen H. Metabolite correlation permutation after mice acute exposure to PM 2.5: Holistic exploration of toxicometabolomics by network analysis. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 352:124128. [PMID: 38729510 DOI: 10.1016/j.envpol.2024.124128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 04/28/2024] [Accepted: 05/07/2024] [Indexed: 05/12/2024]
Abstract
Many environmental toxicants can cause systemic effects, such as fine particulate matter (PM2.5), which can penetrate the respiratory barrier and induce effects in multiple tissues. Although metabolomics has been used to identify biomarkers for PM2.5, its multi-tissue toxicology has not yet been explored holistically. Our objective is to explore PM2.5 induced metabolic alterations and unveil the intra-tissue responses along with inter-tissue communicational effects. In this study, following a single intratracheal instillation of multiple doses (0, 25, and 150 μg as the control, low, and high dose), non-targeted metabolomics was employed to evaluate the metabolic impact of PM2.5 across multiple tissues. PM2.5 induced tissue-specific and dose-dependent disturbances of metabolites and their pathways. The remarkable increase of both intra- and inter-tissue correlations was observed, with emphasis on the metabolism connectivity among lung, spleen, and heart; the tissues' functional specificity has marked their toxic modes. Beyond the inter-status comparison of the metabolite fold-changes, the current correlation network built on intra-status can offer additional insights into how the multiple tissues and their metabolites coordinately change in response to external stimuli such as PM2.5 exposure.
Collapse
Affiliation(s)
- Hao Chang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory & State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen, 361102, PR China
| | - Xi Zhang
- Key Laboratory of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen, 361021, PR China
| | - Zhonghua Lu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory & State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen, 361102, PR China
| | - Biling Gao
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory & State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen, 361102, PR China
| | - Heqing Shen
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory & State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen, 361102, PR China; Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, 361003, PR China.
| |
Collapse
|
48
|
Iftimie S, Gabaldó-Barrios X, Penadés-Nadal J, Canela-Capdevila M, Piñana R, Jiménez-Franco A, López-Azcona AF, Castañé H, Cárcel M, Camps J, Castro A, Joven J. Serum Levels of Arachidonic Acid, Interleukin-6, and C-Reactive Protein as Potential Indicators of Pulmonary Viral Infections: Comparative Analysis of Influenza A, Respiratory Syncytial Virus Infection, and COVID-19. Viruses 2024; 16:1065. [PMID: 39066228 PMCID: PMC11281451 DOI: 10.3390/v16071065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 06/22/2024] [Accepted: 06/26/2024] [Indexed: 07/28/2024] Open
Abstract
Acute respiratory tract infections, including influenza A (FluA), respiratory syncytial virus (RSV) infection, and COVID-19, can aggravate to levels requiring hospitalization, increasing morbidity and mortality. Identifying biomarkers for an accurate diagnosis and prognosis of these infections is a clinical need. We performed a cross-sectional study aimed to investigate the changes in circulating levels of arachidonic acid, interleukin 6 (IL-6), and C-reactive protein (CRP) in patients with FluA, RSV, or COVID-19, and to analyze the potential of these parameters as diagnosis or prognosis biomarkers. We analyzed serum samples from 172 FluA, 80 RSV, and 217 COVID-19 patients, and 104 healthy volunteers. Individuals with lung viral diseases showed reduced arachidonic acid concentrations compared to healthy people, with these differences being most pronounced in the order COVID-19 > RSV > FluA. Conversely, IL-6 and CRP levels were elevated across diseases, with IL-6 emerging as the most promising diagnostic biomarker, with areas under the curve (AUC) of the receiver operating characteristics plot higher than 0.85 and surpassing arachidonic acid and CRP. Moreover, IL-6 displayed notable efficacy in distinguishing between FluA patients who survived and those who did not (AUC = 0.80). These findings may provide useful tools for diagnosing and monitoring the severity of acute viral respiratory tract infections, ultimately improving patient outcomes.
Collapse
Affiliation(s)
- Simona Iftimie
- Department of Internal Medicine, Hospital Universitari de Sant Joan, Institut d’Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43204 Reus, Spain; (S.I.); (J.P.-N.); (A.F.L.-A.); (A.C.)
| | - Xavier Gabaldó-Barrios
- Department of Clinical Laboratory, Hospital Universitari de Sant Joan, Institut d’Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43204 Reus, Spain; (R.P.); (M.C.)
| | - Joan Penadés-Nadal
- Department of Internal Medicine, Hospital Universitari de Sant Joan, Institut d’Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43204 Reus, Spain; (S.I.); (J.P.-N.); (A.F.L.-A.); (A.C.)
| | - Marta Canela-Capdevila
- Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, Institut d’Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43204 Reus, Spain; (M.C.-C.); (A.J.-F.); (H.C.); (J.J.)
| | - Rubén Piñana
- Department of Clinical Laboratory, Hospital Universitari de Sant Joan, Institut d’Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43204 Reus, Spain; (R.P.); (M.C.)
| | - Andrea Jiménez-Franco
- Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, Institut d’Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43204 Reus, Spain; (M.C.-C.); (A.J.-F.); (H.C.); (J.J.)
| | - Ana F. López-Azcona
- Department of Internal Medicine, Hospital Universitari de Sant Joan, Institut d’Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43204 Reus, Spain; (S.I.); (J.P.-N.); (A.F.L.-A.); (A.C.)
| | - Helena Castañé
- Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, Institut d’Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43204 Reus, Spain; (M.C.-C.); (A.J.-F.); (H.C.); (J.J.)
| | - María Cárcel
- Department of Clinical Laboratory, Hospital Universitari de Sant Joan, Institut d’Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43204 Reus, Spain; (R.P.); (M.C.)
| | - Jordi Camps
- Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, Institut d’Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43204 Reus, Spain; (M.C.-C.); (A.J.-F.); (H.C.); (J.J.)
| | - Antoni Castro
- Department of Internal Medicine, Hospital Universitari de Sant Joan, Institut d’Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43204 Reus, Spain; (S.I.); (J.P.-N.); (A.F.L.-A.); (A.C.)
| | - Jorge Joven
- Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, Institut d’Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43204 Reus, Spain; (M.C.-C.); (A.J.-F.); (H.C.); (J.J.)
| |
Collapse
|
49
|
Torrente-Rodríguez RM, Ruiz-Valdepeñas Montiel V, Iftimie S, Montero-Calle A, Pingarrón JM, Castro A, Camps J, Barderas R, Campuzano S, Joven J. Contributing to the management of viral infections through simple immunosensing of the arachidonic acid serum level. Mikrochim Acta 2024; 191:369. [PMID: 38834823 PMCID: PMC11150294 DOI: 10.1007/s00604-024-06440-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 05/15/2024] [Indexed: 06/06/2024]
Abstract
A trendsetting direct competitive-based biosensing tool has been developed and implemented for the determination of the polyunsaturated fatty acid arachidonic acid (ARA), a highly significant biological regulator with decisive roles in viral infections. The designed methodology involves a competitive reaction between the target endogenous ARA and a biotin-ARA competitor for the recognition sites of anti-ARA antibodies covalently attached to the surface of carboxylic acid-coated magnetic microbeads (HOOC-MµBs), followed by the enzymatic label of the biotin-ARA residues with streptavidin-horseradish peroxidase (Strep-HRP) conjugate. The resulting bioconjugates were magnetically trapped onto the sensing surface of disposable screen-printed carbon transducers (SPCEs) to monitor the extent of the biorecognition reaction through amperometry. The operational functioning of the exhaustively optimized and characterized immunosensing bioplatform was highly convenient for the quantitative determination of ARA in serum samples from severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2-) and respiratory syncytial virus (RSV)-infected individuals in a rapid, affordable, trustful, and sensitive manner.
Collapse
Affiliation(s)
- Rebeca M Torrente-Rodríguez
- Departamento de Química Analítica, Facultad de CC. Químicas, Universidad Complutense de Madrid, Pza. de las Ciencias 2, Madrid, 28040, Spain
| | - Víctor Ruiz-Valdepeñas Montiel
- Departamento de Química Analítica, Facultad de CC. Químicas, Universidad Complutense de Madrid, Pza. de las Ciencias 2, Madrid, 28040, Spain
| | - Simona Iftimie
- Servei de Medicina Interna, Hospital Universitari de Sant Joan, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Av. Dr. Josep Laporte 2, Reus, 43204, Spain
| | - Ana Montero-Calle
- Chronic Disease Programme, UFIEC, Instituto de Salud Carlos III, Majadahonda, Madrid, 28220, Spain
| | - José M Pingarrón
- Departamento de Química Analítica, Facultad de CC. Químicas, Universidad Complutense de Madrid, Pza. de las Ciencias 2, Madrid, 28040, Spain
| | - Antoni Castro
- Servei de Medicina Interna, Hospital Universitari de Sant Joan, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Av. Dr. Josep Laporte 2, Reus, 43204, Spain
| | - Jordi Camps
- Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Av. Dr. Josep Laporte 2, Reus, 43204, Spain
| | - Rodrigo Barderas
- Chronic Disease Programme, UFIEC, Instituto de Salud Carlos III, Majadahonda, Madrid, 28220, Spain
- CIBER of Frailty and Healthy Aging (CIBERFES), Madrid, Spain
| | - Susana Campuzano
- Departamento de Química Analítica, Facultad de CC. Químicas, Universidad Complutense de Madrid, Pza. de las Ciencias 2, Madrid, 28040, Spain.
| | - Jorge Joven
- Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Av. Dr. Josep Laporte 2, Reus, 43204, Spain
| |
Collapse
|
50
|
Wang J, Qiu Z, Zeng H, Tan Y, Huang Y, Luo J, Shu W. Long-Term Consumption of Purified Water Altered Amino Acid, Fatty Acid and Energy Metabolism in Livers of Rats. Metabolites 2024; 14:289. [PMID: 38786766 PMCID: PMC11122726 DOI: 10.3390/metabo14050289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 05/16/2024] [Accepted: 05/16/2024] [Indexed: 05/25/2024] Open
Abstract
The consumption of low-mineral water has been increasing worldwide. Drinking low-mineral water is associated with cardiovascular disease, osteopenia, and certain neurodegenerative diseases. However, the specific mechanism remains unclear. The liver metabolic alterations in rats induced by drinking purified water for 3 months were investigated with a metabolomics-based strategy. Compared with the tap water group, 74 metabolites were significantly changed in the purified water group (6 increased and 68 decreased), including 29 amino acids, 11 carbohydrates, 10 fatty acids, 7 short chain fatty acids (SCFAs), and 17 other biomolecules. Eight metabolic pathways were significantly changed, namely aminoacyl-tRNA biosynthesis; nitrogen metabolism; alanine, aspartate and glutamate metabolism; arginine and proline metabolism; histidine metabolism; biosynthesis of unsaturated fatty acids; butanoate metabolism; and glycine, serine and threonine metabolism. These changes suggested that consumption of purified water induced negative nitrogen balance, reduced expression of some polyunsaturated fatty acids and SCFAs, and disturbed energy metabolism in rats. These metabolic disturbances may contribute to low-mineral-water-associated health risks. The health risk of consuming low-mineral water requires attention.
Collapse
Affiliation(s)
- Jia Wang
- Department of Medical English, College of Basic Medicine, Army Medical University, Chongqing 400038, China;
| | - Zhiqun Qiu
- Department of Environmental Hygiene, College of Preventive Medicine, Army Medical University, Chongqin 400038, China; (Z.Q.); (H.Z.); (Y.T.); (Y.H.)
| | - Hui Zeng
- Department of Environmental Hygiene, College of Preventive Medicine, Army Medical University, Chongqin 400038, China; (Z.Q.); (H.Z.); (Y.T.); (Y.H.)
| | - Yao Tan
- Department of Environmental Hygiene, College of Preventive Medicine, Army Medical University, Chongqin 400038, China; (Z.Q.); (H.Z.); (Y.T.); (Y.H.)
| | - Yujing Huang
- Department of Environmental Hygiene, College of Preventive Medicine, Army Medical University, Chongqin 400038, China; (Z.Q.); (H.Z.); (Y.T.); (Y.H.)
| | - Jiaohua Luo
- Department of Environmental Hygiene, College of Preventive Medicine, Army Medical University, Chongqin 400038, China; (Z.Q.); (H.Z.); (Y.T.); (Y.H.)
| | - Weiqun Shu
- Department of Environmental Hygiene, College of Preventive Medicine, Army Medical University, Chongqin 400038, China; (Z.Q.); (H.Z.); (Y.T.); (Y.H.)
| |
Collapse
|