1
|
Helmeczi E, Pandya H, O'Hearn K, McNally D, Britz-McKibbin P. Treatment response variations to a single large bolus of enteral cholecalciferol in vitamin D deficient critically Ill children: Metabolomic insights for precision nutrition. J Steroid Biochem Mol Biol 2025; 250:106720. [PMID: 40064426 DOI: 10.1016/j.jsbmb.2025.106720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 02/02/2025] [Accepted: 03/03/2025] [Indexed: 03/16/2025]
Abstract
Vitamin D deficiency (VDD) is prevalent globally and in pediatric intensive care units, where it represents a modifiable risk factor that may impact patient recovery during hospitalization. Herein, we performed a retrospective analysis of serum samples from a phase-II randomized placebo-controlled trial involving a single large bolus of 10,000 IU/kg vitamin D3 ingested by critically ill children with VDD (25-OH-D < 50 nmol/L). Targeted and untargeted methods were used to comprehensively measure 6 vitamin D metabolites, 239 lipids, 68 polar metabolites, and 4 electrolytes using a multi-step data workflow for compound authentication. Complementary statistical methods classified circulating metabolites/lipids associated with vitamin D repletion following high-dose vitamin D3 intake (n = 20) versus placebo (n = 11) comprising an optional standard of care maintenance dose (< 1000 IU/day). There was a striking increase in median serum concentrations of 25-OH-D3 (4.7-fold), 3-epi-25-OH-D3 (24-fold) and their C3-epimer ratio (6.7-fold) in treated patients on day 3, whereas serum vitamin D3 peaked on day 1 (128-fold) unlike placebo. Treatment response differences were attributed to D3 bioavailability and C3-epimerase activity without evidence of hypercalcemia. For the first time, we report the detection of circulating 3-epi-D3 that was strongly correlated with vitamin D3 uptake (r = 0.898). Metabolomic studies revealed that vitamin D sufficiency (serum 25-OH-D >75 nmol/L) coincided with lower circulating levels of 3-methylhistidine, cystine, S-methylcysteine, uric acid, and two lysophosphatidylcholines 7 days after treatment. Rapid correction of VDD was associated with indicators of lower oxidative stress, inflammation, and muscle protein turn-over that may contribute clinical benefits in high-risk critically ill children.
Collapse
Affiliation(s)
- Erick Helmeczi
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton, ON L8S 4M1, Canada
| | - Haley Pandya
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton, ON L8S 4M1, Canada
| | - Katie O'Hearn
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON K1H 8L1, Canada
| | - Dayre McNally
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON K1H 8L1, Canada; Department of Pediatrics, Children's Hospital of Eastern Ontario, University of Ottawa, Ottawa, ON K1H 8L1, Canada
| | - Philip Britz-McKibbin
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton, ON L8S 4M1, Canada.
| |
Collapse
|
2
|
Liu X, Wei D, Lin J, Dong L, Li Y, Lv M. Plasma lipidome and intracranial aneurysms: A univariable and multivariable Mendelian randomization study. Neuroscience 2025; 573:1-8. [PMID: 40068718 DOI: 10.1016/j.neuroscience.2025.03.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 03/04/2025] [Accepted: 03/07/2025] [Indexed: 03/22/2025]
Abstract
BACKGROUND Recent studies suggest that plasma lipids, including lipoproteins and fatty acids, may contribute to the pathogenesis of intracranial aneurysms (IAs). However, the relationship between a broader range of plasma lipids and IA risk remains unclear. This study uses the Mendelian randomization (MR) approach to explore the causal relationships between 179 plasma lipids and the risk of IAs. METHODS We used summary statistics from a study of 7174 individuals to examine 179 plasma lipids. Data on IAs and aneurysmal subarachnoid hemorrhage (aSAH) were drawn from a study by Bakker MK et al., which included 2070 cases of unruptured IAs, 5 140 cases of aSAH, and 71,934 controls. MR analyses were conducted using inverse variance-weighted as the primary method, with weighted median, weighted mode, and MR-Egger as additional methods. To prioritize lipid risk factors, we applied multivariable Mendelian randomization-Bayesian model averaging. RESULTS We identified 5 plasma lipids associated with IAs and 4 with aSAH. Phosphatidylcholine (14:0_18:2) was significantly associated with both IAs and aSAH, with odds ratios of 1.44 (95 % confidence interval [CI] 1.17-1.77, Padjusted = 0.036) for IAs and 1.53 (95 % CI 1.20-1.94, Padjusted = 0.036) for aSAH. In multivariable MR, phosphatidylcholine (14:0_18:2) and phosphatidylcholine (18:0_20:3) emerged as the strongest risk factors for IAs and aSAH, respectively. CONCLUSION Our study identifies specific plasma lipids, particularly phosphatidylcholine (14:0_18:2) and phosphatidylcholine (18:0_20:3), as significant risk factors for IAs and aSAH. These findings suggest that phosphatidylcholines could serve as predictive biomarkers for aneurysm risk. Further research is needed to validate these associations and clarify the underlying mechanisms.
Collapse
Affiliation(s)
- Xinke Liu
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China; Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Dachao Wei
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Jun Lin
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Linggen Dong
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Youxiang Li
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China; Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
| | - Ming Lv
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China; Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
3
|
Su X, Ding X, Liang J, Zhang L, Zhang Y, Qiao Y, Ma H, Zhang Y, Tang Y, Tan G. Myocardial Lipidomics Revealed Glycerophospholipid and Sphingolipid Metabolism as Therapeutic Targets of Qifu Decoction Against Heart Failure. Biomed Chromatogr 2025; 39:e70063. [PMID: 40110617 DOI: 10.1002/bmc.70063] [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: 01/11/2025] [Revised: 02/20/2025] [Accepted: 03/09/2025] [Indexed: 03/22/2025]
Abstract
Qifu decoction (QFD) has shown potential benefits in treating heart failure. However, the potential mechanism of QFD remains unclear. In this study, myocardial lipidomics, based on ultra-high-performance liquid chromatography coupled with an electrospray ionization hybrid quadrupole Orbitrap mass spectrometry (UPLC-ESI-Q-Exactive/MS), was employed to identify potential therapeutic targets of QFD for treating heart failure in a mice model induced by ligating the left anterior descending coronary artery. It was found that 47 lipid metabolites were associated with heart failure, of which 35 showed a significant reversal during QFD treatment. The QFD-reversed lipid metabolites were mainly located on phosphatidylcholine, lysophosphatidylcholine, sphingomyelin, and ceramide, which were involved in glycerophospholipid and sphingolipid metabolism. The results of Western blotting analysis revealed that QFD could effectively alleviate heart failure through increasing the levels of lysophosphatidylcholine acyltransferase 1 (LPCAT1) and sphingomyelin synthase 1 (SMS1) and reducing the levels of acid sphingomyelinase (aSMase) and phospholipase A2 (PLA2) to regulate the metabolic disorders of glycerophospholipid and sphingolipid metabolism. All these results could be concluded that glycerophospholipid and sphingolipid metabolism were the two crucial target pathways for QFD against heart failure, which laid the theoretical groundwork for its clinical application.
Collapse
Affiliation(s)
- Xuemei Su
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, China
- School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Xin Ding
- School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Junli Liang
- School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Lei Zhang
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, China
- School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Yang Zhang
- School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Yan Qiao
- School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Hongrui Ma
- School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Ya Zhang
- School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Yuping Tang
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Guangguo Tan
- School of Pharmacy, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
4
|
Ma X, Bian W, Song W, Lu Y, Wang Z, Yao Z, Xuan Q. Metabolome profiling across liver lobes and metabolic shifts of the MASLD mice. GENES & NUTRITION 2025; 20:9. [PMID: 40240942 PMCID: PMC12001577 DOI: 10.1186/s12263-025-00768-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 04/04/2025] [Indexed: 04/18/2025]
Abstract
BACKGROUND The mammalian liver executes its vital functions through intricate hepatic biochemistry. However, the complexity of the liver metabolome and its dynamic alterations during metabolic dysfunction-associated steatotic liver disease (MASLD) remain poorly understood. METHODS We established progressive MASLD mouse models through high-fat diet (HFD) and high-fat/high-cholesterol (HFHC) dietary-feeding across multiple time points. Utilizing liquid chromatography-mass spectrometry (LC-MS)-based metabolomics and lipidomics, we systematically mapped the metabolome atlas of the mouse liver across five anatomical segments during the progression of MASLD. RESULTS By integration of data from two assays, we structurally annotated 426 lipids and 118 polar metabolites. The temporal progression of HFD feeding (0, 8, and 16 weeks) resulted in gradual metabolic deterioration across various liver segments. In HFHC-fed mice, metabolic alterations surged sharply from 0 to 8 weeks, followed by moderate progression until 16 weeks in different liver segments. Elevated levels of glycerolipids and cholesteryl esters, along with fluctuating acylcarnitine and fatty acid levels across various liver segments, suggested impaired energy metabolism and disrupted fatty acid oxidation. As MASLD progresses, a shift in sphingolipid metabolism, linked to inflammation, was observed, accompanied by significant alterations in phospholipid turnover patterns. Additionally, amino acid profiles in the livers of HFD-fed and HFHC-fed mice were altered, potentially influencing the regulation of energy metabolism, inflammation, and oxidative stress. These metabolic changes in lipids and amino acids displayed segment-specific patterns, indicating varying sensitivities to inflammation and mitochondrial β-oxidation across different liver lobes. Notably, the left lateral lobe showed heightened sensitivity to metabolic disturbances during MASLD progression. CONCLUSION Our findings provided in-depth understanding in hepatic metabolites of MASLD, offering a comprehensive resource for further investigation.
Collapse
Affiliation(s)
- Xiaolin Ma
- Department of Endocrinology, Shandong Provincial Hospital, Shandong University; Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Jinan, Shandong, 250021, China
- Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, Shandong, 250021, China
- Shandong Institute of Endocrine and Metabolic Diseases, Jinan, Shandong, 250021, China
- "Chuangxin China" Innovation Base of Stem Cell and Gene Therapy for Endocrine Metabolic Diseases, Jinan, Shandong, China
- Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic Diseases, Jinan, Shandong, 250021, China
- Shandong Engineering Research Center of Stem Cell and Gene Therapy for Endocrine and Metabolic Diseases, Jinan, Shandong, 250021, China
| | - Wenbo Bian
- Department of Endocrinology, Shandong Provincial Hospital, Shandong University; Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Jinan, Shandong, 250021, China
- Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, Shandong, 250021, China
- Shandong Institute of Endocrine and Metabolic Diseases, Jinan, Shandong, 250021, China
- "Chuangxin China" Innovation Base of Stem Cell and Gene Therapy for Endocrine Metabolic Diseases, Jinan, Shandong, China
- Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic Diseases, Jinan, Shandong, 250021, China
- Shandong Engineering Research Center of Stem Cell and Gene Therapy for Endocrine and Metabolic Diseases, Jinan, Shandong, 250021, China
| | - Wenting Song
- Department of Endocrinology, Shandong Provincial Hospital, Shandong University; Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Jinan, Shandong, 250021, China
- Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, Shandong, 250021, China
- Shandong Institute of Endocrine and Metabolic Diseases, Jinan, Shandong, 250021, China
- "Chuangxin China" Innovation Base of Stem Cell and Gene Therapy for Endocrine Metabolic Diseases, Jinan, Shandong, China
- Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic Diseases, Jinan, Shandong, 250021, China
- Shandong Engineering Research Center of Stem Cell and Gene Therapy for Endocrine and Metabolic Diseases, Jinan, Shandong, 250021, China
| | - Yitong Lu
- Department of Endocrinology, Shandong Provincial Hospital, Shandong University; Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Jinan, Shandong, 250021, China
- Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, Shandong, 250021, China
- Shandong Institute of Endocrine and Metabolic Diseases, Jinan, Shandong, 250021, China
- "Chuangxin China" Innovation Base of Stem Cell and Gene Therapy for Endocrine Metabolic Diseases, Jinan, Shandong, China
- Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic Diseases, Jinan, Shandong, 250021, China
- Shandong Engineering Research Center of Stem Cell and Gene Therapy for Endocrine and Metabolic Diseases, Jinan, Shandong, 250021, China
| | - Zhen Wang
- Department of Endocrinology, Shandong Provincial Hospital, Shandong University; Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Jinan, Shandong, 250021, China.
- Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, Shandong, 250021, China.
- Shandong Institute of Endocrine and Metabolic Diseases, Jinan, Shandong, 250021, China.
- "Chuangxin China" Innovation Base of Stem Cell and Gene Therapy for Endocrine Metabolic Diseases, Jinan, Shandong, China.
- Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic Diseases, Jinan, Shandong, 250021, China.
- Shandong Engineering Research Center of Stem Cell and Gene Therapy for Endocrine and Metabolic Diseases, Jinan, Shandong, 250021, China.
| | - Zhenyu Yao
- Department of Endocrinology, Shandong Provincial Hospital, Shandong University; Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Jinan, Shandong, 250021, China.
- Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, Shandong, 250021, China.
- Shandong Institute of Endocrine and Metabolic Diseases, Jinan, Shandong, 250021, China.
- "Chuangxin China" Innovation Base of Stem Cell and Gene Therapy for Endocrine Metabolic Diseases, Jinan, Shandong, China.
- Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic Diseases, Jinan, Shandong, 250021, China.
- Shandong Engineering Research Center of Stem Cell and Gene Therapy for Endocrine and Metabolic Diseases, Jinan, Shandong, 250021, China.
| | - Qiuhui Xuan
- Department of Endocrinology, Shandong Provincial Hospital, Shandong University; Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Jinan, Shandong, 250021, China.
- Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, Shandong, 250021, China.
- Shandong Institute of Endocrine and Metabolic Diseases, Jinan, Shandong, 250021, China.
- "Chuangxin China" Innovation Base of Stem Cell and Gene Therapy for Endocrine Metabolic Diseases, Jinan, Shandong, China.
- Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic Diseases, Jinan, Shandong, 250021, China.
- Shandong Engineering Research Center of Stem Cell and Gene Therapy for Endocrine and Metabolic Diseases, Jinan, Shandong, 250021, China.
| |
Collapse
|
5
|
Narasimhan K, Tint MT, Chen L, Mir SA, Sadananthan SA, Michael N, Ramasamy A, Tan KML, Mishra P, Bendt AK, Tan KH, Cameron-Smith D, Chong YS, Gluckman PD, Meikle PJ, Leow MKS, Yap F, Lee YS, Yng CS, Eriksson JG, Velan SS, Karnani N, Wenk MR. Sexual dimorphism in the association of umbilical cord blood lipidome with abdominal fat in early childhood. BMC Med 2025; 23:215. [PMID: 40223079 PMCID: PMC11995507 DOI: 10.1186/s12916-025-04030-0] [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: 06/27/2024] [Accepted: 03/21/2025] [Indexed: 04/15/2025] Open
Abstract
BACKGROUND Although the associations between cord blood lipidome and neonatal birth weight are established, it remains uncertain whether sexual dimorphism in fetal fat accumulation extends to the relationship between cord blood lipid profiles and neonatal abdominal fat compartments. Understanding these relationships could provide insights into early sex-specific differences in lipid metabolism. METHODS We conducted lipidomics of umbilical cord blood plasma samples (350 (46.6%) girls and 401 (53.4%) boys) from the Growing Up in Singapore Towards healthy Outcomes (GUSTO) birth cohort. Abdominal fat compartments-superficial subcutaneous adipose tissue (sSAT), deep SAT (dSAT), and intra-abdominal adipose tissue (IAT)-were quantified by magnetic resonance imaging within 2 weeks of birth in 239 subjects. Linear regression models were used to assess sex differences in lipid species associated with abdominal fat compartments. RESULTS Newborn girls had significantly higher superficial and deep subcutaneous adipose tissue volumes compared to boys, whereas intra-abdominal adipose tissue volumes were similar between sexes. In the pooled analysis, cord blood plasma lipids showed distinct associations with different fat depots: 38 lipid species were associated with sSAT, 4 with dSAT, and 38 with IAT. In sex-stratified analyses, 13 lipids were associated with sSAT in girls and 3 in boys, whereas dSAT showed associations with 45 lipids in boys but none in girls. These sex differences were primarily observed in ether-linked phospholipids and ceramides. Notably, no significant associations were observed between lipids and IAT in either sex, suggesting depot-specific sexual dimorphism in early life. CONCLUSIONS Our study reveals sexual dimorphism in the associations between cord blood lipidome and abdominal adiposity, suggesting depot-specific patterns in adipose tissue development and lipid metabolism in early life.
Collapse
Affiliation(s)
- Kothandaraman Narasimhan
- Institute for Human Development and Potential (IHDP), Agency for Science, Technology and Research (A*STAR), Brenner Centre for Molecular Medicine , 30 Medical Drive, Singapore, 117609, Singapore.
| | - Mya Thway Tint
- Institute for Human Development and Potential (IHDP), Agency for Science, Technology and Research (A*STAR), Brenner Centre for Molecular Medicine , 30 Medical Drive, Singapore, 117609, Singapore
| | - Li Chen
- Institute for Human Development and Potential (IHDP), Agency for Science, Technology and Research (A*STAR), Brenner Centre for Molecular Medicine , 30 Medical Drive, Singapore, 117609, Singapore
- Life Sciences Institute, Singapore Lipidomics Incubator, National University of Singapore, Singapore, Singapore
| | - Sartaj Ahmad Mir
- Life Sciences Institute, Singapore Lipidomics Incubator, National University of Singapore, Singapore, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Suresh Anand Sadananthan
- Institute for Human Development and Potential (IHDP), Agency for Science, Technology and Research (A*STAR), Brenner Centre for Molecular Medicine , 30 Medical Drive, Singapore, 117609, Singapore
| | - Navin Michael
- Institute for Human Development and Potential (IHDP), Agency for Science, Technology and Research (A*STAR), Brenner Centre for Molecular Medicine , 30 Medical Drive, Singapore, 117609, Singapore
| | - Adaikalavan Ramasamy
- Agency for Science, Technology and Research (A*STAR), Genome Institute of Singapore (GIS), 60 Biopolis Street, Genome, Singapore, 138672, Republic of Singapore
| | - Karen Mei-Ling Tan
- Institute for Human Development and Potential (IHDP), Agency for Science, Technology and Research (A*STAR), Brenner Centre for Molecular Medicine , 30 Medical Drive, Singapore, 117609, Singapore
| | - Priti Mishra
- Institute for Human Development and Potential (IHDP), Agency for Science, Technology and Research (A*STAR), Brenner Centre for Molecular Medicine , 30 Medical Drive, Singapore, 117609, Singapore
| | - Anne K Bendt
- Life Sciences Institute, Singapore Lipidomics Incubator, National University of Singapore, Singapore, Singapore
| | - Kok Hian Tan
- Maternal Fetal Medicine, KK Women's and Children's Hospital (KKH), Singapore, Singapore
| | - David Cameron-Smith
- Institute for Human Development and Potential (IHDP), Agency for Science, Technology and Research (A*STAR), Brenner Centre for Molecular Medicine , 30 Medical Drive, Singapore, 117609, Singapore
- Singapore Institute of Food and Biotechnology Innovation (SIFBI), Agency for Science, Technology and Research (A*STAR), 14 Medical Drive, MD6, #07-02, Singapore, Singapore
| | - Yap Seng Chong
- Institute for Human Development and Potential (IHDP), Agency for Science, Technology and Research (A*STAR), Brenner Centre for Molecular Medicine , 30 Medical Drive, Singapore, 117609, Singapore
- Department of Obstetrics and Gynaecology and Human Potential Translational Research programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Peter D Gluckman
- Institute for Human Development and Potential (IHDP), Agency for Science, Technology and Research (A*STAR), Brenner Centre for Molecular Medicine , 30 Medical Drive, Singapore, 117609, Singapore
- Centre for Human Evolution, Adaptation and Disease, Liggins Institute, University of Auckland, Auckland, New Zealand
| | - Peter J Meikle
- Baker Heart and Diabetes Institute AU, Melbourne, VIC, 3004, Australia
- Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Bundoora, VIC, 3086, Australia
| | - Melvin Khee Shing Leow
- Institute for Human Development and Potential (IHDP), Agency for Science, Technology and Research (A*STAR), Brenner Centre for Molecular Medicine , 30 Medical Drive, Singapore, 117609, Singapore
- Singapore Institute of Food and Biotechnology Innovation (SIFBI), Agency for Science, Technology and Research (A*STAR), 14 Medical Drive, MD6, #07-02, Singapore, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Department of Endocrinology, Tan Tock Seng Hospital, Singapore, Singapore
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore, Singapore
| | - Fabian Yap
- Division of Paediatric Medicine, KK Women's and Children's Hospital, Singapore, Singapore
| | - Yung Seng Lee
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Chan Shiao Yng
- Institute for Human Development and Potential (IHDP), Agency for Science, Technology and Research (A*STAR), Brenner Centre for Molecular Medicine , 30 Medical Drive, Singapore, 117609, Singapore
| | - Johan G Eriksson
- Institute for Human Development and Potential (IHDP), Agency for Science, Technology and Research (A*STAR), Brenner Centre for Molecular Medicine , 30 Medical Drive, Singapore, 117609, Singapore
- Department of Obstetrics and Gynaecology and Human Potential Translational Research programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Folkhalsan Research Center, Helsinki, Finland
- Department of General Practice and Primary Health Care, University of Helsinki, Helsinki, Finland
| | - S Sendhil Velan
- Institute for Human Development and Potential (IHDP), Agency for Science, Technology and Research (A*STAR), Brenner Centre for Molecular Medicine , 30 Medical Drive, Singapore, 117609, Singapore
| | - Neerja Karnani
- Institute for Human Development and Potential (IHDP), Agency for Science, Technology and Research (A*STAR), Brenner Centre for Molecular Medicine , 30 Medical Drive, Singapore, 117609, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Clinical Data Engagement, Bioinformatics Institute, Agency for Science Technology and Research (A*STAR), Singapore, Singapore
| | - Markus R Wenk
- Life Sciences Institute, Singapore Lipidomics Incubator, National University of Singapore, Singapore, Singapore.
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar.
| |
Collapse
|
6
|
Cheng Y, Zhao F, Wang J, Luo H, Mao R, Yu Y, Wang Y, Tan J, Hao X, Wang Y. Preparation, activity, and mechanistic insights of processed Polygala tenuifolia glycoprotein in ameliorating Alzheimer's disease. Int J Biol Macromol 2025; 309:143069. [PMID: 40220839 DOI: 10.1016/j.ijbiomac.2025.143069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Revised: 03/25/2025] [Accepted: 04/09/2025] [Indexed: 04/14/2025]
Abstract
This study examined the efficacy and mechanism of Polygala tenuifolia glycoprotein (ZPG) in alleviating Alzheimer's disease (AD) for clinical application. ZPG's effects were tested in scopolamine hydrobromide-induced AD mice using behavioral, histological, and biomarker investigations. Additionally, 16S rDNA sequencing and lipidomics revealed ZPG's impact on gut microbiota and lipid metabolism, supported by pathway enrichment and correlation analyses. JNK pathway modulation was studied in vitro with purified and characterized ZPG-2. Results showed ZPG significantly improved cognitive deficits, reduced hippocampal pathology, and normalized APP, p-JNK, bax, and bcl-2 levels in AD mice. It also modulated gut microbiota and lipid metabolism, particularly glycerophospholipid pathways. ZPG-2 exhibited neuroprotective effects in Aβ25-35-induced PC12 cells by reducing apoptosis, inhibiting LDH release, and regulating oxidative stress and JNK activity. Structural analysis identified ZPG-2 as a 26 kDa glycoprotein with an O-linked glycopeptide bond and random coil conformation. Correlation analysis showed significant gut microbiota-AD biomarker relationships. These findings suggest ZPG may alleviate AD by reducing oxidative stress, inhibiting apoptosis, modulating gut microbiota, enhancing lipid metabolism, and suppressing the JNK signaling pathway. ZPG may be medicinal, however, more research is needed to validate its efficacy and mechanisms. This study lays the foundation for ZPG as an AD therapy for the future.
Collapse
Affiliation(s)
- Yangang Cheng
- Shanxi University of Chinese Medicine, Shanxi, Jinzhong 030619, China
| | - Fuxia Zhao
- Shanxi University of Chinese Medicine, Shanxi, Jinzhong 030619, China
| | - Jing Wang
- Shanxi University of Chinese Medicine, Shanxi, Jinzhong 030619, China
| | - Hong Luo
- The University of Adelaide, Adelaide, SA 5005, Australia
| | - Rui Mao
- Shanxi University of Chinese Medicine, Shanxi, Jinzhong 030619, China
| | - Yuetong Yu
- Shanxi University of Chinese Medicine, Shanxi, Jinzhong 030619, China
| | - Yan Wang
- Shanxi University of Chinese Medicine, Shanxi, Jinzhong 030619, China
| | - Jinyan Tan
- Shanxi University of Chinese Medicine, Shanxi, Jinzhong 030619, China.
| | - Xuliang Hao
- Shanxi Academy of Traditional Chinese Medicine, Shanxi, Taiyuan 030024, China.
| | - Yingli Wang
- Shanxi University of Chinese Medicine, Shanxi, Jinzhong 030619, China.
| |
Collapse
|
7
|
Wu G, Wang M, Du Z, Li Z, Han T, Xie Z, Gu W. Tea polyphenol EGCG enhances the improvements of calorie restriction on hepatic steatosis and obesity while reducing its adverse outcomes in obese rats. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156744. [PMID: 40228342 DOI: 10.1016/j.phymed.2025.156744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 02/24/2025] [Accepted: 04/08/2025] [Indexed: 04/16/2025]
Abstract
BACKGROUND Currently, calorie restriction (CR) is popular among young people as a way to lose weight and prevent obesity. However, CR can also cause a series of side effects, such as weight regain after resuming free eating. Tea polyphenol epigallocatechin-3-gallate (EGCG) has been widely recognized as antiobesity effects. However, whether EGCG can enhance the antiobesity effect of CR and reduce its adverse outcomes is still unclear. PURPOSE This study aimed to explore the enhancing effect and molecular mechanism of EGCG supplementation on CR in improving hepatic steatosis and obesity. METHODS The enhancing effect and molecular mechanism of EGCG supplementation on CR in alleviating hepatic steatosis and obesity were explored using a leptin receptor-knockout (LepR KO) rat model by performing biochemical, histochemistry, qPCR, plasma lipidomic, and gut microbiota analysis. RESULTS Our results showed that CR plus EGCG exhibited enhanced preventive effects in reducing blood glucose, insulin, TC, TG, LDL-C, and FFA levels in plasma, and protection against hepatic steatosis in LepR KO rats than CR alone. In addition, CR plus EGCG remarkably reduced oxidative stress and systemic inflammatory responses in LepR KO rats. Moreover, the combined intervention showed an enhanced improvement effect on the homeostasis of gut microbiota than CR alone, including increasing gut microbiota diversity and modulating microbiota composition. Plasma lipidomics analysis showed that CR plus EGCG significantly improved glycerophospholipid, glycerolipid and sphingolipid metabolism in LepR KO rats. Mechanistic studies showed that CR combined EGCG enhanced SIRT6 and suppressed SREBP1 and FAS expression in the livers of LepR KO rats than CR alone, thereby improving host lipid metabolism. CONCLUSION This study demonstrated that EGCG enhance the improvements of CR on hepatic steatosis and obesity in LepR KO rats, and reduce its adverse outcomes, especially in reducing hepatic lipogenesis and maintaining homeostasis of gut microbiota. This study provides a dietary strategy for preventing weight rebound following the transition from CR to a free diet by supplementing EGCG, suggesting that CR plus EGCG may offer a promising therapy for managing obesity in humans.
Collapse
Affiliation(s)
- Guohuo Wu
- Engineering Technology Research Center of Anti-Aging Chinese Herbal Medicine of Anhui Province, School of Biology and Food Engineering, Fuyang Normal University, Fuyang, China; State Key Laboratory of Tea Plant Biology and Utilization, School of Tea and Food Sciences and Technology, Anhui Agricultural University, Hefei, China
| | - Mengdi Wang
- Department of General Surgery, The First Affiliated Hospital of Fuyang Normal University, Fuyang, China
| | - Zhaofeng Du
- Engineering Technology Research Center of Anti-Aging Chinese Herbal Medicine of Anhui Province, School of Biology and Food Engineering, Fuyang Normal University, Fuyang, China; State Key Laboratory of Tea Plant Biology and Utilization, School of Tea and Food Sciences and Technology, Anhui Agricultural University, Hefei, China
| | - Zhuang Li
- Biotechnology Center, Anhui Agricultural University, Hefei, China
| | - Tingting Han
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea and Food Sciences and Technology, Anhui Agricultural University, Hefei, China
| | - Zhongwen Xie
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea and Food Sciences and Technology, Anhui Agricultural University, Hefei, China.
| | - Wei Gu
- Laboratory Animal Center, Anhui Medical University, Hefei, China; State Key Laboratory of Tea Plant Biology and Utilization, School of Tea and Food Sciences and Technology, Anhui Agricultural University, Hefei, China.
| |
Collapse
|
8
|
Zhao H, Xiao Q, An Y, Wang M, Zhong J. Phospholipid metabolism and drug resistance in cancer. Life Sci 2025; 372:123626. [PMID: 40210119 DOI: 10.1016/j.lfs.2025.123626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 03/27/2025] [Accepted: 04/06/2025] [Indexed: 04/12/2025]
Abstract
Phospholipids, complex lipids prevalent in the human body, play crucial roles in various pathophysiological processes. Beyond their synthesis and degradation, phospholipids can influence chemoresistance by participating in ferroptosis. Extensive evidence highlights the significant link between tumor drug resistance and phospholipids. Therefore, drugs targeting phospholipid metabolism itself or the synthesis of corresponding composite materials will effectively overcome the difficulties of clinical tumor treatment.
Collapse
Affiliation(s)
- Hu Zhao
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China
| | - Qian Xiao
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China; Department of Clinical Laboratory Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China
| | - Yangfang An
- Yiyang Central Hospital, Yiyang, Hunan 413099, PR China
| | - Mu Wang
- Clinical Mass Spectrometry Laboratory, Clinical Research Institute, Affiliated Nanhua Hospital, University of South China, Hengyang, PR China.
| | - Jing Zhong
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China; Institute of Clinical Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China.
| |
Collapse
|
9
|
Ünal Uzun Ö, Eneş D, Çınar M, Günlemez Adugit A, Uçar B, Duranoğlu A, Obuz UB, Çelebier M, Lay İ. Cord blood metabolomic profiling in high risk newborns born to diabetic, obese, and overweight mothers: preliminary report. J Pediatr Endocrinol Metab 2025:jpem-2024-0605. [PMID: 40196912 DOI: 10.1515/jpem-2024-0605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Accepted: 03/23/2025] [Indexed: 04/09/2025]
Abstract
OBJECTIVES Newborns of diabetic and obese/overweight mothers face long-term metabolic risks. Untargeted cord blood metabolomic analysis using quadrupole time-of-flight liquid chromatography/mass spectrometry (Q-TOF LC/MS) was performed to explore metabolic alterations and pathways in these high-risk infants. METHODS Cord blood samples were collected from 46 newborns born to mothers with gestational diabetes (10), obesity (14), overweight (18), type 2 diabetes mellitus (3), type 1 diabetes mellitus (1), and 20 newborns born to healthy mothers. Q-TOF LC/MS was used to investigate the alterations in cord blood metabolomic profiles. Data processing was conducted using MZmine 2.53. Putative metabolites were idendtified using MetaboAnalyst 6.0. RESULTS Distinct metabolite profiles were observed between diabetes and control groups. Significant identical trend in 19 metabolites were determined in both diabetes and obesity + overweight group vs. control group. Key pathways included steroid and bile acid biosynthesis. Upregulated oxidative stress, clues to sphingophospholipid metabolism, high levels of dihomo-gamma-linolenic acid (DGLA), pantothenic acid, and TRH were detected. The kynurenine pathway was prominent in the diabetes group. CONCLUSIONS Estrogen metabolites from the 16- and 2-pathways may indicate metabolic risk, with increased downstream flux under diabetic conditions. Accelerated bile acid synthesis may alter fetal metabolic programming, since bile acids play crucial roles in cellular energy regulation and signaling. Elevated pantothenic acid, essential for the production of coenzyme-A, suggests significant alterations in carbohydrate, protein, and fat metabolism. High serum DGLA levels emerge as a potential biomarker for metabolic abnormalities. Increased plasma kynurenines could predict cardiovascular risks. Larger targeted studies are required to validate these metabolic profiles and pathways.
Collapse
Affiliation(s)
- Özlem Ünal Uzun
- Department of Pediatrics, Division of Metabolism, Faculty of Medicine, Kocaeli University, Kocaeli, Türkiye
| | - Duygu Eneş
- Department of Analytical Chemistry, Faculty of Pharmacy, Başkent University, Ankara, Türkiye
| | - Müge Çınar
- Department of Pediatrics, Division of Metabolism, Faculty of Medicine, Kocaeli University, Kocaeli, Türkiye
| | - Ayla Günlemez Adugit
- Department of Pediatrics, Division of Neonatology, Faculty of Medicine, Kocaeli University, Kocaeli, Türkiye
| | - Büşra Uçar
- Department of Analytical Chemistry, Faculty of Pharmacy, Hacettepe University, Ankara, Türkiye
| | - Ali Duranoğlu
- Department of Pediatrics, Faculty of Medicine, Kocaeli University, Kocaeli, Türkiye
| | - Ufuk Bozkurt Obuz
- Scientific and Technological Research Center, Sinop University, Sinop, Türkiye
| | - Mustafa Çelebier
- Department of Analytical Chemistry, Faculty of Pharmacy, Hacettepe University, Ankara, Türkiye
| | - İncilay Lay
- Department of Medical Biochemistry, Hacettepe University, Ankara, Türkiye
- Central Laboratory, Hacettepe University Hospitals, Ankara, Türkiye
| |
Collapse
|
10
|
Muratoğlu B, Özdemir C, Eylem CC, Reçber T, Nemutlu E, Alpdündar-Bulut E, Vargel İ, Uçkan-Çetinkaya D. Detailed characterization of bone marrow adipose tissue mesenchymal stem cells in healthy donor, Fanconi anemia, and acute myeloid leukemia. Bone 2025; 193:117413. [PMID: 39894290 DOI: 10.1016/j.bone.2025.117413] [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: 11/20/2024] [Revised: 01/08/2025] [Accepted: 01/27/2025] [Indexed: 02/04/2025]
Abstract
Bone marrow is a complex tissue featuring distinct cellular organization and diverse cell types. Bone marrow adipose tissue (BMAT) is a dynamic component crucial for tissue function and disease processes. This study explores differences between bone marrow-derived mesenchymal stem cells (BM-MSCs) and BMAT-derived mesenchymal stem cells (BMAT-MSCs), isolated from the same cavity, examining their differentiation potential and secretory profiles. BM-MSCs and BMAT-MSCs both exhibit classical mesenchymal characteristics, with over 90 % positivity for markers such as CD105 and CD29. Notably, BMAT-MSCs display significantly higher differentiation potential than BM-MSCs, with enhanced osteogenic and adipogenic capabilities, as indicated by increased calcium accumulation and lipid storage. In Fanconi anemia (FA) and acute myeloid leukemia (AML), osteogenic potential is limited, indicating impaired differentiation under these pathological conditions. Gene expression analysis of adipogenic molecules and metabolic regulators revealed significant differences in expression profile between BM- and BMAT-MSCs, particularly during adipogenic differentiation, indicating distinct characteristics that were more notable in FAs and AMLs. Furthermore, metabolomic profiling of BM plasma, using GC-MS for in-vivo niche reflection, and lipid analysis via LC-qTOF-MS show significant lipidomic alterations in patient samples, highlighting metabolic dysregulation and lipid remodeling. Lipid-mediated signaling and membrane composition changes appear integral to disease mechanisms. In conclusion, this study highlights the distinctive molecular and metabolomic profiles and adaptive mechanisms of BM- and BMAT-MSCs in bone marrow pathologies.
Collapse
Affiliation(s)
- Bihter Muratoğlu
- Institute of Health Sciences, Department of Stem Cell Sciences and Center for Stem Cell Research and Development (PEDI-STEM), Hacettepe University, Ankara, Türkiye; Institute of Health Sciences, Department of Stem Cell Sciences, Hacettepe University, Ankara, Türkiye
| | - Cansu Özdemir
- Institute of Health Sciences, Department of Stem Cell Sciences and Center for Stem Cell Research and Development (PEDI-STEM), Hacettepe University, Ankara, Türkiye; Institute of Health Sciences, Department of Stem Cell Sciences, Hacettepe University, Ankara, Türkiye.
| | - Cemil Can Eylem
- Department of Analytical Chemistry, Hacettepe University, Ankara, Türkiye
| | - Tuba Reçber
- Department of Analytical Chemistry, Hacettepe University, Ankara, Türkiye
| | - Emirhan Nemutlu
- Department of Analytical Chemistry, Hacettepe University, Ankara, Türkiye
| | - Esin Alpdündar-Bulut
- Institute of Health Sciences, Department of Stem Cell Sciences and Center for Stem Cell Research and Development (PEDI-STEM), Hacettepe University, Ankara, Türkiye; Division of Hematology, Department of Pediatrics, Hacettepe University, Ankara, Türkiye
| | - İbrahim Vargel
- Faculty of Medicine, Department of Plastic, Reconstructive and Aesthetic Surgery, Hacettepe University, Ankara, Türkiye
| | - Duygu Uçkan-Çetinkaya
- Institute of Health Sciences, Department of Stem Cell Sciences and Center for Stem Cell Research and Development (PEDI-STEM), Hacettepe University, Ankara, Türkiye; Institute of Health Sciences, Department of Stem Cell Sciences, Hacettepe University, Ankara, Türkiye; Division of Hematology, Department of Pediatrics, Hacettepe University, Ankara, Türkiye
| |
Collapse
|
11
|
Huang SY, Yang ZJ, Cheng J, Li HY, Chen S, Huang ZH, Chen JD, Xiong RG, Yang MT, Wang C, Li MC, Song S, Huang WG, Wang DL, Li HB, Lan QY. Choline alleviates cognitive impairment in sleep-deprived young mice via reducing neuroinflammation and altering phospholipidomic profile. Redox Biol 2025; 81:103578. [PMID: 40056720 PMCID: PMC11930228 DOI: 10.1016/j.redox.2025.103578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 02/12/2025] [Accepted: 02/27/2025] [Indexed: 03/10/2025] Open
Abstract
Cognitive impairment resulting from insufficient sleep poses a significant public health concern, particularly in children. The effects and mechanisms of choline on cognitive impairment caused by sleep deprivation are unknown. Chronic sleep deprivation is induced in young mice in this study, followed by feeding diet containing 11.36 g/kg choline bitartrate. Choline supplementation significantly improves spatial learning ability. Functional MRI results reveal the hippocampus as a key region affected by sleep deprivation, where choline supplementation notably preserves hippocampal structural integrity and enhanced connectivity. Additionally, choline ameliorates hippocampal pathological injury, reduces blood-brain barrier permeability and serum brain injury biomarkers. Choline also reduces inflammation and oxidative stress biomarkers, and mitigates microglial activation in the hippocampus, which preserves synaptic plasticity. A key finding is the changes of hippocampal phospholipidomic profile along with cognitive function, and a total of 313 phospholipid molecules are identified. Choline increases the levels of total phospholipid and sub-classes (particularly PC), which are strongly correlated with reduced neuroinflammation and oxidative stress biomarkers, as well as improved cognitive outcomes. Furthermore, there are similar findings in some phospholipid molecules such as PC 36:1, PC O-33:0, PC p-38:3, PE 36:3, PE p-42:4 and PS 44:12. These findings highlight that choline alleviates cognitive impairment in sleep deprivation via reducing neuroinflammation and oxidative stress as well as altering phospholipidomic profile. This study suggests that choline could develop into functional food or medicine ingredient to prevent and treat cognitive impairment by sleep disturbances, particularly children and adolescents.
Collapse
Affiliation(s)
- Si-Yu Huang
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Zhi-Jun Yang
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Jin Cheng
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Hang-Yu Li
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Si Chen
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Zi-Hui Huang
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Jie-Dong Chen
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Ruo-Gu Xiong
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Meng-Tao Yang
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Chen Wang
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Meng-Chu Li
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Shuang Song
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing, 100050, China
| | - Wen-Ge Huang
- Center of Experimental Animals, Sun Yat-sen University, Guangzhou, 510080, China
| | - Dong-Liang Wang
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Hua-Bin Li
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China.
| | - Qiu-Ye Lan
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China; School of Public Health, Guangdong Pharmaceutical University, Guangzhou, 510006, China.
| |
Collapse
|
12
|
Bobin P, Mitanchez D, Castellano B, Grit I, Moyon T, Raux A, Vambergue A, Winer N, Darmaun D, Michel C, Le Drean G, Alexandre-Gouabau MC. A specific metabolomic and lipidomic signature reveals the postpartum resolution of gestational diabetes mellitus or its evolution to type 2 diabetes in rat. Am J Physiol Endocrinol Metab 2025; 328:E493-E512. [PMID: 39947887 DOI: 10.1152/ajpendo.00396.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 11/11/2024] [Accepted: 02/01/2025] [Indexed: 04/01/2025]
Abstract
Gestational diabetes mellitus (GDM) represents a major public health concern due to adverse maternal postpartum and long-term outcomes. Current strategies to manage GDM fail to reduce the maternal risk to develop later impaired glucose tolerance (IGT) and type 2 diabetes (T2D). In a rodent model of diet-induced GDM without obesity, we explored the perinatal metabolic adaptations in dams with gestational IGT followed by either persistent or resolved postpartum IGT. Female Sprague-Dawley rats were fed a high-fat high-sucrose (HFHS) or a chow [control group (CTL)] diet, 1 wk before mating and throughout gestation (G). Following parturition, HFHS dams were randomized to two subgroups: one switched to a chow diet and the other one maintained on an HFHS diet throughout lactation (L). Oral glucose tolerance tests (OGTTs) were performed, and plasma metabolome-lipidome were characterized at G12 and L12. We found that 1) in GDM-pregnant dams, IGT was associated with incomplete fatty acid oxidation (FAO), enhanced gluconeogenesis, altered insulin signaling, and oxidative stress; 2) improved glucose tolerance postpartum seemed to restore complete FAO along with elevation of nervonic acid-containing sphingomyelins, assumed to impart β-cell protection; and 3) persistence of IGT after delivery was associated with metabolites known to predict the early onset of insulin and leptin resistance, with maintained liver dysfunction. Our findings shed light on the impact of postpartum IGT evolution on maternal metabolic outcome after an episode of GDM. They suggest innovative strategies, implemented shortly after delivery and targeted on these biomarkers, should be explored to curb or delay the transition from GDM to T2D in these mothers.NEW & NOTEWORTHY Specific metabolomic/lipidomic features are associated with GDM postpartum outcomes. GDM-pregnant dams exhibit partial fatty acid oxidation and boosted gluconeogenesis. Resolution of postpartum IGT relies on nervonic acid-sphingomyelin, a β-cell protector. Postpartum IGT persistence suggests muscle insulin resistance and liver dysfunction.
Collapse
Affiliation(s)
- Paul Bobin
- Nantes Université, INRAE, UMR1280 PhAN, Nantes, France
| | - Delphine Mitanchez
- Department of Neonatology, Bretonneau Hospital, François Rabelais University, Tours, France
- INSERM UMRS_938, Centre de Recherche Saint Antoine, Paris, France
| | | | - Isabelle Grit
- Nantes Université, INRAE, UMR1280 PhAN, Nantes, France
| | - Thomas Moyon
- Nantes Université, INRAE, UMR1280 PhAN, Nantes, France
| | - Axel Raux
- Oniris, INRAE, LABERCA, Nantes, France
| | - Anne Vambergue
- Department of Diabetology, Hospital Huriez, CHRU de Lille, University of Lille, EGID-UMR 8199, Lille, France
| | - Norbert Winer
- Nantes Université, INRAE, UMR1280 PhAN, Nantes, France
- Department of Obstetrics and Gynecology, CHU, Nantes University Hospital, Nantes, France
| | | | | | | | | |
Collapse
|
13
|
Zhang Q, Huang J, Kang P, Su G. Biotransformation of bis(2-chloroethyl)2-chloroethylphosphphonate, and its effects on metabolism of lipid molecules in primary mouse hepatocytes. CHEMOSPHERE 2025; 374:144195. [PMID: 39938318 DOI: 10.1016/j.chemosphere.2025.144195] [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: 11/25/2024] [Revised: 01/27/2025] [Accepted: 02/03/2025] [Indexed: 02/14/2025]
Abstract
Bis(2-chloroethyl)2-chloroethylphosphphonate (B2CE2CEPP) was recently reported as a contaminant in underground water samples, however, information regarding its biotransformation and adverse effects is relatively rare. In this study, B2CE2CEPP and tris(2-chloroethyl) phosphate (TCEP; a typical organophosphate triester with a very similar structure as compared to B2CE2CEPP) were used as target contaminants to comparatively investigate their cytotoxicity, biotransformation, and alteration of lipid metabolism by use of an in vitro primary hepatocyte assay of ICR mice. This study aims to thoroughly investigate the cytotoxicity, biotransformation, and lipid metabolism effects of B2CE2CEPP and TCEP on primary hepatocytes of ICR mice, in order to reveal the potential health risks of these two organophosphorus compounds. The results showed that both B2CE2CEPP and TCEP were cytotoxic with LC50 values of 456 μM (B2CE2CEPP) and 1250 μM (TCEP), respectively. Both compounds underwent significant metabolic transformation follow exposure to primary mouse hepatocytes (PMHs), and a total of 4 potential metabolites were identified based on high-resolution orbitrap mass spectrometry technique. Exposure to TCEP and B2CE2CEPP led to lipid metabolism disorders in PMHs. TG (16:0/16:1/18:1) showed high sensitivity to B2CE2CEPP exposure and exhibited a significant upregulation trend, making it a potential biomarker significantly influenced by B2CE2CEPP exposure. Lipidomic analysis showed that B2CE2CEPP affected lipid metabolic pathways including thiamine metabolism, one carbon pool by folate, and folatesteroid hormone biosynthesis. The present study filled a knowledge gap in the current research on the biotransformation and adverse effects of B2CE2CEPP, and emphasized the importance on consistent monitoring of B2CE2CEPP and its structure-like compounds in various environmental samples.
Collapse
Affiliation(s)
- Qi Zhang
- Key Laboratory of Environmental Remediation and Ecological Health, Ministry of Industry and Information Technology, School of Environmental and Biological Engineering, Nanjing University of Science and Technology, Nanjing, 210094, China
| | - Jianan Huang
- Key Laboratory of Environmental Remediation and Ecological Health, Ministry of Industry and Information Technology, School of Environmental and Biological Engineering, Nanjing University of Science and Technology, Nanjing, 210094, China
| | - Pingping Kang
- Key Laboratory of Environmental Remediation and Ecological Health, Ministry of Industry and Information Technology, School of Environmental and Biological Engineering, Nanjing University of Science and Technology, Nanjing, 210094, China
| | - Guanyong Su
- Key Laboratory of Environmental Remediation and Ecological Health, Ministry of Industry and Information Technology, School of Environmental and Biological Engineering, Nanjing University of Science and Technology, Nanjing, 210094, China.
| |
Collapse
|
14
|
Li Q, Chao T, Wang Y, He P, Zhang L, Wang J. Metabolomics and transcriptomics analyses reveal the complex molecular mechanisms by which the hypothalamus regulates sexual development in female goats. BMC Genomics 2025; 26:303. [PMID: 40148778 PMCID: PMC11951529 DOI: 10.1186/s12864-025-11492-2] [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: 12/12/2024] [Accepted: 03/17/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND The hypothalamus is a critical organ that regulates sexual development in animals. However, current research on the hypothalamic regulation of sexual maturation in female goats remains limited. In this study, we conducted metabolomic and transcriptomic analyses on the hypothalamic tissues of female Jining grey goats at different stages of sexual development (1 day old (neonatal, D1, n = 5), 2 months old (prepuberty, M2, n = 5), 4 months old (sexual maturity, M4, n = 5), and 6 months old (breeding period, M6, n = 5)). RESULTS A total of 418 differential metabolites (DAMs) were identified in this study, among which the abundance of metabolites such as anserine, L-histidine, carnosine, taurine, and 4-aminobutyric gradually increased with the progression of sexual development. These metabolites may regulate neuronal development and hormone secretion processes by influencing the metabolism of histidine and phenylalanine. Through combined transcriptomic and metabolomic analyses, we identified that differentially expressed genes such as mitogen-activated protein kinase kinase kinase 9 (MAP3K9), prune homolog 2 with BCH domain (PRUNE2), and potassium voltage-gated channel interacting protein 4(KCNIP4) may jointly regulate the development and energy metabolism of hypothalamic Gonadotropin-releasing hormone neurons in conjunction with DAMs, including LPC22:5, 2-Arachidonyl Glycerol ether, LPE22:5, and Lysops22:5. Additionally, we elucidated the molecular mechanism through which glutathione metabolism regulates sexual maturation in goats. CONCLUSIONS In summary, this study illustrates the dynamic changes in metabolites and mRNA within hypothalamic tissue during postnatal sexual maturation in female Jining grey goats. This research may provide significant scientific insights for future animal breeding.
Collapse
Affiliation(s)
- Qing Li
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an City, 271014, Shandong Province, China
- Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Shandong Agricultural University, Tai'an City, 271014, Shandong Province, China
| | - Tianle Chao
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an City, 271014, Shandong Province, China.
- Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Shandong Agricultural University, Tai'an City, 271014, Shandong Province, China.
| | - Yanyan Wang
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an City, 271014, Shandong Province, China
- Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Shandong Agricultural University, Tai'an City, 271014, Shandong Province, China
| | - Peipei He
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an City, 271014, Shandong Province, China
- Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Shandong Agricultural University, Tai'an City, 271014, Shandong Province, China
| | - Lu Zhang
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an City, 271014, Shandong Province, China
- Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Shandong Agricultural University, Tai'an City, 271014, Shandong Province, China
| | - Jianmin Wang
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an City, 271014, Shandong Province, China.
- Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Shandong Agricultural University, Tai'an City, 271014, Shandong Province, China.
| |
Collapse
|
15
|
Tan F, Zheng Y, Wang C, Huang J, Liu X, Su W, Chen X, Yang Z. Effects of Chenpi Jiaosu on serum metabolites and intestinal microflora in a dyslipidemia population: a randomized controlled pilot trial. Front Endocrinol (Lausanne) 2025; 16:1552117. [PMID: 40225325 PMCID: PMC11985429 DOI: 10.3389/fendo.2025.1552117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Accepted: 02/10/2025] [Indexed: 04/15/2025] Open
Abstract
Introduction Dyslipidemia is a critical risk factor for atherosclerosis and cardiovascular/cerebrovascular events, necessitating effective long-term management. However, conventional lipid-lowering drugs such as statins and fibrates are limited by adverse effects, including hepatotoxicity and myopathy, which restrict their prolonged use. Traditional Chinese medicine (TCM) and natural health products offer potential alternatives with multi-target mechanisms and improved safety profiles. Tangerine Peel Enzyme Drink (CPJS), a fermented health product derived from tangerine peel, has demonstrated lipid-modulating properties. This study aimed to evaluate the efficacy and safety of CPJS in improving dyslipidemia and explore its underlying metabolic and microbiological mechanisms. Methods A randomized, double-blind, parallel-controlled clinical trial was conducted with 72 participants (55 completers). Participants were divided into CPJS and control groups, receiving an 8-week intervention. Primary outcomes included changes in body weight and serum triglycerides (TG), while safety was assessed via liver/kidney function, creatine kinase, blood, and urine tests. Serum metabolomics (93 differential metabolites identified) and intestinal microbiota analysis were performed to elucidate metabolic pathways and microbial shifts. KEGG enrichment analysis mapped metabolites to biological pathways, such as lipid and amino acid metabolism. Results The CPJS group exhibited significant reductions in body weight and TG levels post-intervention (p < 0.05), with no adverse effects observed in safety biomarkers. Metabolomic profiling revealed alterations in fatty acyl, glycerophospholipid, and organic acid metabolites, indicating CPJS modulates lipid metabolism and energy homeostasis. KEGG analysis linked these changes to pathways including triglyceride degradation and amino acid metabolism. Additionally, CPJS increased specific gut microbial taxa associated with lipid regulation, suggesting a microbiome-mediated mechanism. Discussion CPJS demonstrates efficacy in improving dyslipidemia through dual mechanisms: direct modulation of triglyceride metabolism and indirect regulation via gut microbiota. Its safety profile aligns with findings from natural products like Cyclocarya paliurus and tempeh, which mitigate lipid abnormalities without hepatotoxicity. The multi-target action of CPJS mirrors TCM principles, where compounds like quercetin and flavonoids in CPJS may synergistically inhibit cholesterol synthesis and enhance lipid clearance. However, further research is needed to isolate active components and validate microbial contributions. Compared to synthetic drugs, CPJS offers a safer adjunct therapy, addressing limitations of current pharmacotherapies. Future studies should explore dose-response relationships and long-term outcomes in diverse populations.
Collapse
Affiliation(s)
- Fei Tan
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yuying Zheng
- Guangdong Engineering Research Center of Commercialization of Medical Institution Preparations and Traditional Chinese Medicines, Engineering Technology Research Center of Commercialization of Linnan Special Medical Institution Preparations, Experimental Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong Clinical Research Academy of Chinese Medicine, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong Engineering Research Center of Commercialization of Medical Institution Preparations and Traditional Chinese Medicines, Guangzhou, China
- Guangdong Engineering Technology Research Center of Commercialization of Linnan Special Medical Institution Preparations, Guangzhou, China
| | - Chengcheng Wang
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jiaying Huang
- The TCM Department of Longyuan Daguan Community Health Service Center, Shenzhen Longgang Orthopaedics Hospital, Shenzhen, China
| | - Xin Liu
- Production department, Guangzhou Baiyunshan Guanghua Pharmaceutical co, LTD, Guangzhou, China
| | - Weiwei Su
- School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Xinyan Chen
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhimin Yang
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
16
|
Yan W, Shi X, Zhao Y, Liu X, Jia X, Gao L, Yuan J, Liao A, Yasui H, Wang X, Wang X, Zhang R, Wang H. Microbiota-reprogrammed phosphatidylcholine inactivates cytotoxic CD8 T cells through UFMylation via exosomal SerpinB9 in multiple myeloma. Nat Commun 2025; 16:2863. [PMID: 40128181 PMCID: PMC11933704 DOI: 10.1038/s41467-025-57966-5] [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: 04/17/2024] [Accepted: 03/09/2025] [Indexed: 03/26/2025] Open
Abstract
Gut microbiome influences tumorigenesis and tumor progression through regulating the tumor microenvironment (TME) and modifying blood metabolites. However, the mechanisms by which gut microbiome and blood metabolites regulate the TME in multiple myeloma (MM) remain unclear. By employing16S rRNA gene sequencing coupled with metagenomics and ultra-performance liquid chromatography coupled with quadrupole time-of-flight mass spectrometry, we find that Lachnospiraceae are high and phosphatidylcholine (PC) are low in MM patients. We further show that Lachnospiraceae inhibits PC production from MM cells and enhances cytotoxic CD8 T cell function. Mechanistically, PC promotes Sb9 mRNA maturation in MM cells by LIN28A/B via lysophosphatidic acid, thus enhances exosamal Sb9 production. Exosamal Sb9 then reduces GZMB expression by suppressing tumor protein p53 (TP53) UFMylation via the competitive binding of TP53 with the ubiquitin-fold modifier conjugating enzyme 1 in CD8 T cells. We thus show that Lachnospiraceae and PC may be potential therapeutic targets for MM treatment.
Collapse
Affiliation(s)
- Wei Yan
- Department of Hematology, Shengjing Hospital, China Medical University, Shenyang, China
| | - Xue Shi
- Department of Hematology, Shengjing Hospital, China Medical University, Shenyang, China
| | - Yun Zhao
- Department of Radiology, Shengjing Hospital, China Medical University, Shenyang, China
| | - Xiaoyu Liu
- Department of Hematology, Shengjing Hospital, China Medical University, Shenyang, China
| | - Xueming Jia
- Department of Hematology, Shengjing Hospital, China Medical University, Shenyang, China
| | - Le Gao
- Department of Hematology, Shengjing Hospital, China Medical University, Shenyang, China
| | - Jiahe Yuan
- Department of Hematology, Shengjing Hospital, China Medical University, Shenyang, China
| | - Aijun Liao
- Department of Hematology, Shengjing Hospital, China Medical University, Shenyang, China
| | - Hiroshi Yasui
- Department of Hematology and Oncology, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Xiaobin Wang
- Center for Reproductive Medicine, Shengjing Hospital, China Medical University, Shenyang, China.
| | - Xiaotian Wang
- Department of Urology, Shengjing Hospital, China Medical University, Shenyang, China.
| | - Rui Zhang
- Department of Hematology, The First Affiliated Hospital, China Medical University, Shenyang, China.
| | - Huihan Wang
- Department of Hematology, Shengjing Hospital, China Medical University, Shenyang, China.
| |
Collapse
|
17
|
Wang N, Li H, Wu H, Xia Z, Ren D, Zhang Y, Zhao Y, Zhang H, Zhuang K, Yi L. Untargeted metabolomics combined with lipidomics revealed the effects of myocardial infarction and exercise rehabilitation on blood circulation metabolism of patients based on liquid chromatography-mass spectrometry. J Pharm Biomed Anal 2025; 255:116651. [PMID: 39742691 DOI: 10.1016/j.jpba.2024.116651] [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: 10/07/2024] [Revised: 12/04/2024] [Accepted: 12/22/2024] [Indexed: 01/04/2025]
Abstract
Myocardial infarction (MI) is a major cause of death worldwide. Exercise rehabilitation (ER) is a powerful tool to improve life quality and prognosis of MI patients. Herein, we developed an untargeted metabolomics combined with lipidomics method to qualitatively and quantitatively detect metabolites in plasma. A total of 475 metabolites were annotated according to MS, MS/MS, and quantified by internal standard method. Moreover, medical statistical methods combined with chemometrics were used for metabolomics data mining and interpretation of clinical issues (matched Cohort 1, n = 90, Cohort 2, n = 6). The results illustrated that abnormal lipid metabolism is the most significant metabolic disorder for MI patients. And, three metabolic pathways, bile secretion, HIF-1 signaling pathway, and glutathione metabolism were uncovered in MI patients. Furthermore, glutamine, Phenylacetylglutamine (PAGln) and lysophosphatidylcholine (LPCs) were revealed as the essential biomarkers for ER of MI patients. Our findings revealed the metabolic landscape of MI and metabolic alterations after ER, will underlay potential applications of plasma metabolites in the detection of MI and optimization of ER program.
Collapse
Affiliation(s)
- Na Wang
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming, 650500, China
| | - Huimin Li
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming, 650500, China
| | - Hao Wu
- Faculty of Chemical Engineering, Kunming University of Science and Technology, Kunming, 650500, China; Department of Cardiology, First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650000, China
| | - Zilin Xia
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming, 650500, China
| | - Dabing Ren
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming, 650500, China
| | - Yunmei Zhang
- Department of Cardiology, First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650000, China
| | - Yan Zhao
- Department of Cardiology, First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650000, China
| | - Hong Zhang
- Department of Cardiology, First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650000, China
| | - Ke Zhuang
- Department of Cardiology, First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650000, China.
| | - Lunzhao Yi
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming, 650500, China; Faculty of Chemical Engineering, Kunming University of Science and Technology, Kunming, 650500, China.
| |
Collapse
|
18
|
Mu C, Shao K, Su M, Guo Y, Qiu Y, Sun R, Sun S, Sun Y, Liu C, Wang W, Qin X, Tang C. Lysophosphatidylcholine promoting α-Synuclein aggregation in Parkinson's disease: disrupting GCase glycosylation and lysosomal α-Synuclein degradation. NPJ Parkinsons Dis 2025; 11:47. [PMID: 40089519 PMCID: PMC11910603 DOI: 10.1038/s41531-025-00902-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 03/06/2025] [Indexed: 03/17/2025] Open
Abstract
In Parkinson's Disease (PD), elevated serum lysophosphatidylcholine (LPC) levels correlate with disease progression. However, the mechanisms by which abnormal LPC elevation contributes to PD-related neurotoxicity remain poorly understood. This study aims to investigate the pathogenic role of LPC in dopaminergic neuronal damage and elucidates its underlying mechanisms. Our results showed LPC induces α-synuclein aggregation, exacerbating cognitive dysfunction. LPC activates Cleaved-Caspase3 via the orphan receptor GPR35-ERK signaling pathway, inhibits GRASP65 expression, and disrupts the polarized structure of the Golgi apparatus. This disruption impairs glycosylation and function of glucocerebrosidase (GCase), preventing its transport to lysosomes and leading to glucosylceramide (GlcCer) accumulation, a scaffold for α-synuclein aggregation. LPC also disrupts the autophagolysosomal pathway and lysosomal acidification, exacerbating toxic α-synuclein accumulation. Restoring GCase glycosylation, limiting GlcCer synthesis, or blocking ERK signaling mitigates these effects. This study highlights LPC's role in promoting α-synuclein aggregation and autophagolysosomal dysfunction, advancing our understanding of PD pathology.
Collapse
Affiliation(s)
- Chunyan Mu
- Department of Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Kaiquan Shao
- Department of Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Mingyu Su
- Department of Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Yurong Guo
- Department of Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Yuxiang Qiu
- Department of Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Ruiao Sun
- Department of Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Sihan Sun
- Department of Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Yaoyu Sun
- Department of Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Chenkai Liu
- Department of Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Wei Wang
- The Second School of Clinical Medicine, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
| | - Xiaoling Qin
- Department of Neurology, Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Fudan University, Shanghai, 200031, China.
| | - Chuanxi Tang
- Department of Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
- The Research and Engineering Center of Xuzhou neurodegenerative disease diagnosis and treatment biologics, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
| |
Collapse
|
19
|
Mou Y, Tang Y, Zheng X, Liu X, Wu X, Wang H, Zeng J, Rao Q, Ben-David Y, Li Y, Huang L. Unraveling the molecular mechanisms of Fufangduzhong formula in alleviating high-fat diet-induced non-alcoholic fatty liver disease in mice. Front Pharmacol 2025; 16:1542143. [PMID: 40144651 PMCID: PMC11936930 DOI: 10.3389/fphar.2025.1542143] [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: 12/09/2024] [Accepted: 02/19/2025] [Indexed: 03/28/2025] Open
Abstract
Background Non-alcoholic fatty liver disease (NAFLD) is a common chronic liver disease, characterized by hepatic lipid accumulation. The Fufangduzhong formula (FFDZ) is a traditional Chinese medicine (TCM) formulation composed of Eucommia ulmoides Oliv., Leonurus artemisia (Lour.) S. Y. Hu, Prunella vulgaris Linn, Uncariarhynchophylla (Miq.) Miq. ex Havil., and Scutellaria baicalensis Georgi. It has demonstrated hepatoprotective effects and the ability to reduce lipid accumulation. However, its mechanisms against NAFLD remain unclear. Methods UPLC-MS/MS was used to identify FFDZ metabolites. C57BL/6J mice were fed a high-fat diet (HFD) supplemented with or without FFDZ (HFD+L, 0.45 g/kg/d; HFD+H, 0.9 g/kg/d) for 12 weeks. Biochemical indicators and histopathological observations were utilized to assess the extent of metabolic homeostasis disorder and hepatic steatosis. An analysis of differentially expressed genes and regulated signaling pathways was conducted using hepatic transcriptomics. Metabolomics analysis was performed to investigate the significantly changed endogenous metabolites associated with NAFLD in mice serum using UPLC-Q-TOF/MS. Western blot was employed to detect proteins involved in the lipid metabolism-related signaling pathways. Oleic acid-induced hepatic steatosis was used to examine the lipid-lowering effect of FFDZ-containing serum in vitro. Results A total of eight active metabolites were identified from the FFDZ formula and FFDZ-containing serum through UPLC-MS/MS analysis. FFDZ reduced body weight, liver weight, and levels of inflammatory cytokines, and it ameliorated hepatic steatosis, serum lipid profiles, insulin sensitivity, and glucose tolerance in mice with HFD-induced NAFLD. Transcriptomics revealed that FFDZ modulated the lipid metabolism-related pathways, including the PPAR signaling pathway, Fatty acid metabolism, and AMPK signaling pathway. Meanwhile, Western blot analysis indicated that FFDZ downregulated the expression of lipid synthesis-related proteins (Srebp-1c, Acly, Scd-1, Fasn, Acaca, and Cd36) and upregulated the fatty acid oxidation-related proteins (p-Ampk, Ppar-α, and Cpt-1). Furthermore, metabolomics identified FFDZ-mediated reversal of phospholipid dysregulation (PC, PE, LPC, LPE). Additionally, FFDZ-containing serum remarkedly reduced OA-induced lipid accumulation in HepG2 cells. Conclusion The present results demonstrate that FFDZ exerts anti-NAFLD effects by enhancing glucose tolerance and insulin sensitivity, as well as regulating the Ampk signaling pathway to ameliorate lipid metabolism disorder, lipotoxicity, hepatic steatosis, and inflammatory responses.
Collapse
Affiliation(s)
- Yu Mou
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Guizhou Medical University, Guiyang, China
- Natural Products Research Center of Guizhou Province, Guiyang, China
- School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
| | - Yao Tang
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Guizhou Medical University, Guiyang, China
- Natural Products Research Center of Guizhou Province, Guiyang, China
- School of Basic Medical, Guizhou Medical University, Guiyang, China
| | - Xiuyan Zheng
- Guizhou Institute of Integrated Agriculture Development, Guiyang, China
| | - Xiang Liu
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Guizhou Medical University, Guiyang, China
- Natural Products Research Center of Guizhou Province, Guiyang, China
- School of Basic Medical, Guizhou Medical University, Guiyang, China
| | - Xuemei Wu
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Guizhou Medical University, Guiyang, China
- Natural Products Research Center of Guizhou Province, Guiyang, China
- School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
| | - Hongji Wang
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Guizhou Medical University, Guiyang, China
- Natural Products Research Center of Guizhou Province, Guiyang, China
- School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
| | - Jie Zeng
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Guizhou Medical University, Guiyang, China
- Natural Products Research Center of Guizhou Province, Guiyang, China
- School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
| | - Qing Rao
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Guizhou Medical University, Guiyang, China
- Natural Products Research Center of Guizhou Province, Guiyang, China
- School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
| | - Yaacov Ben-David
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Guizhou Medical University, Guiyang, China
- Natural Products Research Center of Guizhou Province, Guiyang, China
- School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
- School of Basic Medical, Guizhou Medical University, Guiyang, China
| | - Yanmei Li
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Guizhou Medical University, Guiyang, China
- Natural Products Research Center of Guizhou Province, Guiyang, China
- School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
| | - Lei Huang
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Guizhou Medical University, Guiyang, China
- Natural Products Research Center of Guizhou Province, Guiyang, China
- School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
| |
Collapse
|
20
|
Sivanandham S, Sivanandham R, Xu C, Symmonds J, Sette P, He T, Funderburg N, Abdel-Mohsen M, Landay A, Apetrei C, Pandrea I. Plasma lipidomic alterations during pathogenic SIV infection with and without antiretroviral therapy. Front Immunol 2025; 16:1475160. [PMID: 40129985 PMCID: PMC11931036 DOI: 10.3389/fimmu.2025.1475160] [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: 08/02/2024] [Accepted: 02/05/2025] [Indexed: 03/26/2025] Open
Abstract
Introduction Lipid profiles change in human immunodeficiency virus (HIV) infection and correlate with inflammation. Lipidomic alterations are impacted by multiple non-HIV-related behavioral risk factors; thus, use of animal models in which these behavioral factors are controlled may inform on the specific lipid changes induced by simian immunodeficiency virus (SIV) infection and/or antiretroviral therapy (ART). Methods Using ultrahigh Performance Liquid Chromatography-Tandem Mass Spectroscopy, we assessed and compared (ANOVA) longitudinal lipid changes in naïve and ART-treated SIV-infected pigtailed macaques (PTMs). Key parameters of infection (IL-6, TNFa, D-dimer, CRP and CD4+ T cell counts) were correlated (Spearman) with lipid concentrations at critical time points of infection and treatment. Results Sphingomyelins (SM) and lactosylceramides (LCER) increased during acute infection, returning to baseline during chronic infection; Hexosylceramides (HCER) increased throughout infection, being normalized with prolonged ART; Phosphatidylinositols (PI) and lysophosphatidylcholines (LPC) decreased with SIV infection and did not return to normal with ART; Phosphatidylethanolamines (PE), lysophosphatidylethanolamines (LPE) and phosphatidylcholines (PC) were unchanged by SIV infection, yet significantly decreased throughout ART. Specific lipid species (SLS) were also substantially modified by SIV and/or ART in most lipid classes. In conclusion, using a metabolically controlled model, we identified specific lipidomics signatures of SIV infection and/or ART, some of which were similar to people living with HIV (PWH). Many SLS were identical to those involved in development of organ dysfunctions encountered in virally suppressed individuals. Lipid changes also correlated with markers of disease progression, inflammation and coagulation. Discussion Our data suggest that lipidomic profile alterations contribute to residual systemic inflammation and comorbidities seen in HIV/SIV infections and therefore may be used as biomarkers of SIV/HIV comorbidities. Further exploration into the benefits of interventions targeting dyslipidemia is needed for the prevention HIV-related comorbidities.
Collapse
Affiliation(s)
- Sindhuja Sivanandham
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
- Division of Infectious Diseases, Department of Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Ranjit Sivanandham
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
- Division of Infectious Diseases, Department of Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Cuiling Xu
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
- Division of Infectious Diseases, Department of Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Jen Symmonds
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Infectious Diseases and Microbiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, United States
| | - Paola Sette
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
- Division of Infectious Diseases, Department of Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Tianyu He
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
- Division of Infectious Diseases, Department of Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Nicholas Funderburg
- Division of Medical Laboratory Science, School of Health and Rehabilitation Sciences, The Ohio State University, Columbus, OH, United States
| | - Mohamed Abdel-Mohsen
- Vaccine and Immunotherapy Center, Wistar Institute, Philadelphia, PA, United States
| | - Alan Landay
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX, United States
| | - Cristian Apetrei
- Division of Infectious Diseases, Department of Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Infectious Diseases and Microbiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, United States
| | - Ivona Pandrea
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Infectious Diseases and Microbiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
21
|
Perpiñá-Clérigues C, Mellado S, Galiana-Roselló C, García-García F, Pascual M. Unraveling the Impact of TLR4 and Sex on Chronic Alcohol Consumption-Induced Lipidome Dysregulation in Extracellular Vesicles. J Proteome Res 2025; 24:1197-1208. [PMID: 39907520 DOI: 10.1021/acs.jproteome.4c00786] [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: 02/06/2025]
Abstract
The lipids that form extracellular vesicles (EVs) play critical structural and regulatory roles, and cutting-edge bioinformatics strategies have shown the ability to decipher lipid metabolism and related molecular mechanisms. We previously demonstrated that alcohol abuse induces an inflammatory immune response through Toll-like receptor 4 (TLR4), leading to structural and cognitive dysfunction. This study evaluated how TLR4 and sex as variables (male/female) impact the lipidome of plasma-resident EVs after chronic alcohol exposure. Using a mouse model of chronic ethanol exposure in wild-type and TLR4-deficient mice, enrichment networks generated by LINEX2 highlighted significant ethanol-induced changes in the EV lipid substrate-product of enzyme reactions associated with glycerophospholipid metabolism. We also demonstrated ethanol-induced differences in Lipid Ontology enrichment analysis in EVs, focusing on terms related to lipid bilayer properties. A lipid abundance analysis revealed higher amounts of significant lipid subclasses in all experimental comparisons associated with inflammatory responses and EV biogenesis/secretion. These findings suggest that interrogating EV lipid abundance with a sensitive lipidomic-based strategy can provide deep insight into the molecular mechanisms underlying biological processes associated with sex, alcohol consumption, and TLR4 immune responses and open new avenues for biomarker identification and therapeutic development.
Collapse
Affiliation(s)
- Carla Perpiñá-Clérigues
- Computational Biomedicine Laboratory, Príncipe Felipe Research Center (CIPF), C/ Eduardo Primo Yúfera, 3, 46012 Valencia, Spain
- Department of Physiology, School of Medicine and Dentistry, University of Valencia, 46010 Valencia, Spain
| | - Susana Mellado
- Department of Physiology, School of Medicine and Dentistry, University of Valencia, 46010 Valencia, Spain
| | - Cristina Galiana-Roselló
- Computational Biomedicine Laboratory, Príncipe Felipe Research Center (CIPF), C/ Eduardo Primo Yúfera, 3, 46012 Valencia, Spain
| | - Francisco García-García
- Computational Biomedicine Laboratory, Príncipe Felipe Research Center (CIPF), C/ Eduardo Primo Yúfera, 3, 46012 Valencia, Spain
| | - María Pascual
- Department of Physiology, School of Medicine and Dentistry, University of Valencia, 46010 Valencia, Spain
| |
Collapse
|
22
|
Bastos JM, Colaço B, Baptista R, Gavina C, Vitorino R. Innovations in heart failure management: The role of cutting-edge biomarkers and multi-omics integration. JOURNAL OF MOLECULAR AND CELLULAR CARDIOLOGY PLUS 2025; 11:100290. [PMID: 40129519 PMCID: PMC11930597 DOI: 10.1016/j.jmccpl.2025.100290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 02/11/2025] [Accepted: 02/27/2025] [Indexed: 03/26/2025]
Abstract
Heart failure (HF) remains a major cause of morbidity and mortality worldwide and represents a major challenge for diagnosis, prognosis and treatment due to its heterogeneity. Traditional biomarkers such as BNP and NT-proBNP are valuable but insufficient to capture the complexity of HF, especially phenotypes such as HF with preserved ejection fraction (HFpEF). Recent advances in multi-omics technology and novel biomarkers such as cell-free DNA (cfDNA), microRNAs (miRNAs), ST2 and galectin-3 offer transformative potential for HF management. This review explores the integration of these innovative biomarkers into clinical practice and highlights their benefits, such as improved diagnostic accuracy, enhanced risk stratification and non-invasive monitoring capabilities. By leveraging multi-omics approaches, including lipidomics and metabolomics, clinicians can uncover new pathways, refine the classification of HF phenotypes, and develop personalized therapeutic strategies tailored to individual patient profiles. Remarkable advances in proteomics and metabolomics have identified biomarkers associated with key HF mechanisms such as mitochondrial dysfunction, inflammation and fibrosis, paving the way for targeted therapies and early interventions. Despite the promising results, significant challenges remain in translating these findings into routine care, including high costs, technical limitations and the need for large-scale validation studies. This report argues for an integrative, multi-omics-based model to overcome these obstacles and emphasizes the importance of collaboration between researchers, clinicians and policy makers. By linking innovative science with practical applications, multi-omics approaches have the potential to redefine HF management and lead to better patient outcomes and more sustainable healthcare systems.
Collapse
Affiliation(s)
- Jose Mesquita Bastos
- Department of Medical Sciences, Institute of Biomedicine iBiMED, University of Aveiro, 3810-193 Aveiro, Portugal
- Cardiology Department, Hospital Infante D. Pedro, Centro Hospitalar do Baixo Vouga, Aveiro, Portugal
| | - Beatriz Colaço
- Department of Medical Sciences, Institute of Biomedicine iBiMED, University of Aveiro, 3810-193 Aveiro, Portugal
- Cardiology Department, Hospital Infante D. Pedro, Centro Hospitalar do Baixo Vouga, Aveiro, Portugal
| | - Rui Baptista
- Department of Cardiology, Centro Hospitalar de Entre o Douro e Vouga, Santa Maria da Feira, Portugal
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
| | - Cristina Gavina
- Pedro Hispano Hospital - ULS Matosinhos, Matosinhos, Portugal
- Cardiology Department, Faculty of Medicine, University of Porto, Oporto, Portugal
- RISE- Health Research Network, Faculty of Medicine, University of Porto, Oporto, Portugal
| | - Rui Vitorino
- Department of Medical Sciences, Institute of Biomedicine iBiMED, University of Aveiro, 3810-193 Aveiro, Portugal
- Cardiovascular R&D Centre - UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine of the University of Porto, 4200-319 Porto, Portugal
| |
Collapse
|
23
|
Wu W, Huynh K, Du JC, She G, Duong T, Ziemann M, Zhao WB, Deng XL, Meikle PJ, Du XJ. Hippo pathway activation causes multiple lipid derangements in a murine model of cardiomyopathy. Biochim Biophys Acta Mol Cell Biol Lipids 2025; 1870:159590. [PMID: 39709046 DOI: 10.1016/j.bbalip.2024.159590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 12/02/2024] [Accepted: 12/17/2024] [Indexed: 12/23/2024]
Abstract
Metabolic reprogramming occurs in cardiomyopathy and heart failure contributing to progression of the disease. Activation of cardiac Hippo pathway signaling has been implicated in mediating mitochondrial dysfunction and metabolic reprogramming in cardiomyopathy, albeit influence of Hippo pathway on lipid profile is unclear. Using a dual-omics approach, we determined alterations of cardiac lipids in a mouse model of cardiomyopathy due to enhanced Hippo signaling and explored molecular mechanisms. Lipidomic profiling discovered multiple alterations in lipid classes, notably reduction of triacylglycerol, diacylglycerol, phospholipids and ether lipids, and elevation of sphingolipids and lysophosphatidylcholine. Mechanistically, we found downregulated expression of PPARα and PGC-1α at mRNA and protein levels, and downregulated expression of PPARα-target genes, indicating attenuated transcriptional activity of PPARα/PGC-1α. Lipidomics-guided transcriptomic analysis revealed dysregulated expression of gene sets that were responsible for enhanced biosynthesis of ceramides, suppression of TG biosynthesis, storage, hydrolysis and mitochondrial fatty acid oxidation, and reduction of peroxisome-localized biosynthesis of ether lipids. Collectively, Hippo pathway activation with attenuated PPARα/PGC-1α signaling is the underlying mechanism for alterations in cardiac lipids in cardiomyopathy and failing heart.
Collapse
Affiliation(s)
- Wei Wu
- Department of Cardiology, Shaanxi Provincial Hospital, Xi'an, China; Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Kevin Huynh
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Jin-Chan Du
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Gang She
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Thy Duong
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Mark Ziemann
- School of Life and Environmental Sciences, Deakin University, Geelong, Victoria, Australia; Bioinformatics Working Group, Burnet Institute, Melbourne, Victoria, Australia
| | - Wei-Bo Zhao
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; Department of Endocrinology, The Ninth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Xiu-Ling Deng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Peter J Meikle
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia.
| | - Xiao-Jun Du
- Department of Cardiology, Shaanxi Provincial Hospital, Xi'an, China; Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an Jiaotong University Health Science Center, Xi'an, China; Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia.
| |
Collapse
|
24
|
Ren J, Yu L, Lin J, Liu Y, Ma L, Huang Y, Sun N, Deng Y, Zhong D, Zhou B, Jiang B, Yan M. Elevated 18:1 lysophosphatidylcholine contributes to neuropathic pain in peripheral nerve injury. Reg Anesth Pain Med 2025:rapm-2024-106195. [PMID: 40016127 DOI: 10.1136/rapm-2024-106195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 02/10/2025] [Indexed: 03/01/2025]
Abstract
BACKGROUND Neuropathic pain is a maladaptive and chronic condition with limited effective treatments. Although recent studies have suggested that certain lipid metabolites, like lysophosphatidylcholine (LPC), may contribute to chronic pain, their specific roles and mechanisms remain unclear. OBJECTIVE This study investigated the role and mechanism of LPC(18:1), a lipid subtype, in neuropathic pain caused by nerve injury. METHODS Using a mouse model of spinal nerve ligation, LPC(18:1) levels were measured in serum, dorsal root ganglion (DRG), spinal cord (SC) and cerebrospinal fluid (CSF). Nociception was assessed using von Frey and Hargreaves' methods, while molecular analyses explored inflammatory pathways and oxidative stress. RESULTS LPC(18:1) levels significantly increased in the serum, DRG and CSF after nerve injury. Administration of LPC(18:1) induced heightened pain responses and activated inflammatory pathways, including protein kinase C (PKC) and extracellular regulated protein kinase (ERK) in the DRG, as well as glial cells in the SC. The findings suggested that oxidative stress played a role in LPC(18:1) production, and its effects were mediated by G protein-coupled receptor 132 (GPR132). CONCLUSION LPC(18:1) may serve as a potential biomarker and therapeutic target for managing neuropathic pain.
Collapse
Affiliation(s)
- Jinxuan Ren
- Department of Anesthesiology, Zhejiang University School of Medicine Second Affiliated Hospital, Hangzhou, Zhejiang, China
| | - Lina Yu
- Department of Anesthesiology, Zhejiang University School of Medicine Second Affiliated Hospital, Hangzhou, Zhejiang, China
| | - Jiaqi Lin
- Department of Anesthesiology and Pain Management, Shanghai East Hospital, Shanghai, China
| | - Ying Liu
- Department of Anesthesiology, Zhejiang University School of Medicine Second Affiliated Hospital, Hangzhou, Zhejiang, China
| | - Longfei Ma
- Department of Anesthesiology, Zhejiang University School of Medicine Second Affiliated Hospital, Hangzhou, Zhejiang, China
| | - Yangyuxin Huang
- Department of Anesthesiology, Zhejiang University School of Medicine Second Affiliated Hospital, Hangzhou, Zhejiang, China
| | - Na Sun
- Department of Anesthesiology, Zhejiang University School of Medicine Second Affiliated Hospital, Hangzhou, Zhejiang, China
| | - Yutao Deng
- Department of Anesthesiology, Zhejiang University School of Medicine Second Affiliated Hospital, Hangzhou, Zhejiang, China
| | - Da Zhong
- Department of Anesthesiology, Zhejiang University School of Medicine Second Affiliated Hospital, Hangzhou, Zhejiang, China
| | - Binglin Zhou
- Department of Anesthesiology, Zhejiang University School of Medicine Second Affiliated Hospital, Hangzhou, Zhejiang, China
| | - Baochun Jiang
- Department of Anesthesiology, Zhejiang University School of Medicine Second Affiliated Hospital, Hangzhou, Zhejiang, China
| | - Min Yan
- Department of Anesthesiology, Zhejiang University School of Medicine Second Affiliated Hospital, Hangzhou, Zhejiang, China
| |
Collapse
|
25
|
Wang M, Yang T, Xiang Y, Pang J, Wang Y, Sun D. Coix Seed Extract Attenuates Glycolipid Metabolism Disorder in Hyperlipidemia Mice Through PPAR Signaling Pathway Based on Metabolomics and Network Pharmacology. Foods 2025; 14:770. [PMID: 40077474 PMCID: PMC11899454 DOI: 10.3390/foods14050770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Revised: 02/19/2025] [Accepted: 02/20/2025] [Indexed: 03/14/2025] Open
Abstract
Hyperlipidemia is characterized by a high level of blood lipid which poses a serious threat to human health. Coix seed is a traditional crop of medicine and food homology with a wide range of pharmacological actions. To make clear the attenuation effect of coix seed against hyperlipidemia, low and high doses of coix seed extract (CSE) were orally administered to hyperlipidemia model mice developed by high-fat diet (HFD). Our results showed that CSE notably improved liver pathological injury, and oxidative stress, and declined the levels of glucose and lipid in hyperlipidemia mice. Liver metabolomics showed that lipid-related metabolites notably decreased, and pathways of glycolipid metabolism were seriously affected by CSE intervention. Moreover, 16S rRNA sequencing revealed that CSE treatment notably increased the diversity of gut microbiota. Meanwhile, the microbiota with the function of regulating intestinal balance as well as relieving obesity and nervous diseases significantly enhanced while harmful flora notably decreased after CSE intervention. The results of network pharmacology and molecular docking indicated that the PPAR signaling pathway may be the core path of anti-hyperlipidemia for coix seeds. RT-qPCR further verified that the expression levels of genes from the PPAR pathway notably changed by CSE treatment with fat synthesis genes significantly decreased while lipolysis genes notably enhanced. Therefore, coix seed might be a potential candidate for the treatment of hyperlipidemia.
Collapse
Affiliation(s)
- 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; (M.W.); (T.Y.); (Y.X.); (Y.W.)
| | - 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; (M.W.); (T.Y.); (Y.X.); (Y.W.)
| | - Yongjing Xiang
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang 561113, China; (M.W.); (T.Y.); (Y.X.); (Y.W.)
| | - Junxiao Pang
- College of Food Science and Engineering, Guiyang University, Guiyang 550005, China;
| | - Yao Wang
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang 561113, China; (M.W.); (T.Y.); (Y.X.); (Y.W.)
| | - 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; (M.W.); (T.Y.); (Y.X.); (Y.W.)
| |
Collapse
|
26
|
Grzymala B, Þorsteinsson H, Halldórsdóttir DÞ, Sveinsdóttir HS, Sævarsdóttir BR, Norton WHJ, Parker MO, Rolfsson Ó, Karlsson KÆ. Metabolomic and lipidomic profiling reveals convergent pathways in attention deficit hyperactivity disorder therapeutics: Insights from established and emerging treatments. J Pharmacol Exp Ther 2025; 392:103403. [PMID: 40081232 DOI: 10.1016/j.jpet.2025.103403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 02/09/2025] [Indexed: 03/15/2025] Open
Abstract
Attention deficit hyperactivity disorder (ADHD) is a neurodevelopmental disorder with unclear pathological mechanisms. ADHD is treated with both stimulant and nonstimulant medications, but their therapeutic mechanisms and impact on brain metabolites are not fully understood. This study employed an untargeted metabolomics approach with liquid chromatography mass spectrometry to investigate the pathogenesis of ADHD, as well as the effects of established and novel therapeutics. We characterized the metabolomic signatures of the adgrl3.1 mutant zebrafish ADHD model and examined the impact of methylphenidate, guanfacine, atomoxetine, and 5 novel putative therapeutics identified in a prior screen, including amlodipine. Our analysis revealed that the drugs commonly affect pathways related to amino acid and lipid metabolism, specifically involving glycine, serine, threonine, phenylalanine, lysophosphatidylcholine, and sphingomyelin. This convergence on similar metabolic targets was unexpected and suggests a broader, systemic effect of ADHD therapeutics, challenging the traditional view of distinct drug mechanisms. Amlodipine exhibited metabolic effects consistent with established treatments, indicating its potential as a viable alternative or adjunct therapy. These findings provide new insights into the metabolic underpinnings of ADHD and highlight potential targets for developing improved therapeutic strategies. SIGNIFICANCE STATEMENT: This study explores the metabolic pathways affected by attention deficit hyperactivity disorder treatments using a zebrafish adgrl3.1 mutant model. Untargeted metabolomics revealed that both established and novel attention deficit hyperactivity disorder medications influence common amino acid and lipid metabolism pathways, suggesting systemic effects. Notably, amlodipine showed similar impacts as current drugs, offering promise as an alternative therapy.
Collapse
Affiliation(s)
- Bartosz Grzymala
- 3Z, Reykjavik, Iceland; Department of Engineering, Reykjavik University, Reykjavik, Iceland
| | | | | | | | | | - William H J Norton
- Department of Genetics and Genome Biology, University of Leicester, Leicester, United Kingdom
| | - Matthew O Parker
- Surrey Sleep Research Centre, School of Biosciences, University of Surrey, Guildford, United Kingdom
| | - Óttar Rolfsson
- Center for Systems Biology, University of Iceland, Reykjavik, Iceland; Medical Department, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Karl Ægir Karlsson
- 3Z, Reykjavik, Iceland; Department of Engineering, Reykjavik University, Reykjavik, Iceland; Medical Department, School of Health Sciences, University of Iceland, Reykjavik, Iceland.
| |
Collapse
|
27
|
Newsom OJ, Sullivan LB. Defined media reveals the essential role of lipid scavenging to support cancer cell proliferation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.12.637975. [PMID: 40027810 PMCID: PMC11870423 DOI: 10.1101/2025.02.12.637975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Fetal bovine serum (FBS) is a nearly ubiquitous, yet undefined additive in mammalian cell culture media whose functional contributions to promoting cell proliferation remain poorly understood. Efforts to replace serum supplementation in culture media have been hindered by an incomplete understanding of the environmental requirements fulfilled by FBS in culture. Here, we use a combination of live-cell imaging and liquid chromatography-mass spectrometry to elucidate the role of serum in supporting proliferation. We show that serum provides consumed factors that enable proliferation and demonstrate that the serum metal and lipid components are crucial to sustaining proliferation in culture. Importantly, despite access to a wide range of lipid classes, albumin-bound lipids are the primary species consumed during cancer cell proliferation. Furthermore, we find that combinations of the additive ITS, containing necessary metals, and albumin-associated lipid classes are sufficient to replace FBS in culture media. We show that serum-free media enables sensitive quantification of lipid consumption dynamics during cell proliferation, which indicate that fatty acids (FA) are consumed through a mass-action mechanism, with minimal competition from other lipid classes. Finally, we find that pharmacologic disruption of FA activation and incorporation into the cellular lipidome reduces uptake from the environment and impairs cell proliferation. This work therefore identifies metabolic contributions of serum in cell culture settings and provides a framework for building cell culture systems that sustain cell proliferation without the variable and undefined contributions of FBS.
Collapse
Affiliation(s)
- Oliver J. Newsom
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Lucas B. Sullivan
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| |
Collapse
|
28
|
Henze E, Burkhardt RN, Fox BW, Schwertfeger TJ, Gelsleichter E, Michalski K, Kramer L, Lenfest M, Boesch JM, Lin H, Schroeder FC, Kawate T. ATP-release pannexin channels are gated by lysophospholipids. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.10.23.563601. [PMID: 37961151 PMCID: PMC10634739 DOI: 10.1101/2023.10.23.563601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
In addition to its role as cellular energy currency, adenosine triphosphate (ATP) serves as an extracellular messenger that mediates diverse cell-to-cell communication. Compelling evidence supports that ATP is released from cells through pannexins, a family of membrane proteins that form heptameric large-pore channels. However, the activation mechanisms that trigger ATP release by pannexins remain poorly understood. Here, we discover lysophospholipids as endogenous pannexin activators, using activity-guided fractionation of mouse tissue extracts combined with untargeted metabolomics and electrophysiology. We show that lysophospholipids directly and reversibly activate pannexins in the absence of other proteins. Secretomics experiments reveal that lysophospholipid-activated pannexin 1 leads to the release of not only ATP but also other signaling metabolites, such as 5'-methylthioadenosine, which is important for immunomodulation. We also demonstrate that lysophospholipids activate endogenous pannexin 1 in human monocytes, leading to the release of IL-1β through inflammasome activation. Our results provide a connection between lipid metabolism and purinergic signaling, both of which play major roles in immune responses.
Collapse
|
29
|
Volat F, Medhi R, Maggs LZ, Deken MA, Price A, Andrews L, Clark J, Taylor D, Carruthers A, Taylor-Smith E, Pacheco N, Rudge SA, Fraser A, Lopez-Clavijo AF, Sousa BC, Johnson Z, Di Conza G, van der Veen L, Shah P, Sandig H, Sharpe HJ, Farrow S. Pancreatic CAF-Derived Autotaxin Drives Autocrine CTGF Expression to Modulate Protumorigenic Signaling. Mol Cancer Ther 2025; 24:230-241. [PMID: 39570650 DOI: 10.1158/1535-7163.mct-23-0522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 07/26/2024] [Accepted: 11/13/2024] [Indexed: 11/22/2024]
Abstract
Autotaxin (ATX), encoded by ENPP2, is a clinical target in pancreatic ductal adenocarcinoma (PDAC). ATX catalyzes the production of lysophosphatidic acid (LPA), an important regulator within the tumor microenvironment (TME), yet the protumorigenic action of the ATX/LPA axis in PDAC remains unclear. In this study, by interrogating patient samples and cell line datasets, we show that the PDAC TME, rather than cancer cells, is responsible for the majority of ENPP2 expression and highlight a key role for cancer-associated fibroblast (CAF)-derived ATX in autocrine and paracrine protumorigenic signaling. Using the clinical-stage ATX inhibitor, IOA-289, we identified connective tissue growth factor (CTGF), also known as CCN2, as a downstream mediator of ATX signaling in the PDAC CAF-derived cell line, 0082T. Genetic ablation or pharmacologic inhibition of ATX in 0082T CAFs reduced CTGF secretion via modulation of LPA/LPA receptor signaling. Despite the loss of ATX function, extracellular levels of LPA were paradoxically increased, indicating a role for ATX beyond its enzymatic activity and suggesting a role for its LPA chaperone function in the LPA/LPA receptor signaling in CAFs. As CAFs are the main source for CTGF in the PDAC TME, these findings suggest a role for ATX in promoting a protumorigenic microenvironment via modulation of CAF secretion not only via its LPA-producing activity but also via its LPA chaperone function, providing a potential mechanism for the antitumor effects of ATX inhibition.
Collapse
Affiliation(s)
- Fanny Volat
- Cancer Research Horizons, Cambridge, United Kingdom
| | - Ragini Medhi
- Cancer Research Horizons, Cambridge, United Kingdom
| | - Lauren Z Maggs
- Cancer Research Horizons, Cambridge, United Kingdom
- University of Cambridge, Cambridge, United Kingdom
- The Babraham Institute, Cambridge, United Kingdom
| | | | - Alice Price
- Cancer Research Horizons, Cambridge, United Kingdom
| | | | | | - Diane Taylor
- The Babraham Institute, Cambridge, United Kingdom
| | | | | | | | | | - Amy Fraser
- Cancer Research Horizons, Cambridge, United Kingdom
| | | | | | | | | | | | - Pritom Shah
- Cancer Research Horizons, Cambridge, United Kingdom
| | | | | | | |
Collapse
|
30
|
Ducatez F, Berger MG, Pilon C, Plichet T, Lesueur C, Berger J, Belmatoug N, Marret S, Bekri S, Tebani A. Deciphering metabolic shifts in Gaucher disease type 1: a multi-omics study. J Mol Med (Berl) 2025; 103:187-203. [PMID: 39738845 DOI: 10.1007/s00109-024-02512-x] [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: 05/14/2024] [Revised: 11/13/2024] [Accepted: 12/22/2024] [Indexed: 01/02/2025]
Abstract
Gaucher disease (GD), an autosomal recessive lysosomal disorder, primarily affects the lysosomal enzyme β-glucocerebrosidase (GCase), leading to glucosylceramide accumulation in lysosomes. GD presents a wide spectrum of clinical manifestations. This study deploys immune-based proteomics and mass spectrometry-based metabolomics technologies to comprehensively investigate the biochemical landscape in 43 deeply phenotyped type 1 GD patients compared to 59 controls. Conventional and systems biology approaches have been used to analyze the data. The results show promising biological imprints. Elevated phosphatidylcholines in GD patients suggest altered lipid metabolism, potentially due to their increased synthesis. This points to endoplasmic reticulum stress and impaired lipid trafficking, commonly seen in lysosomal diseases. GD patients exhibit an inflammatory profile with elevated cytokines and autoimmune-like inflammation, even in treated patients, highlighting the complexity of GD-related immune imbalances. Mitochondrial dysfunction clues are found through increased oxidative stress markers and altered acylcarnitine profiles in GD patients, suggesting mitochondrial membrane dysfunction affecting carnitine-carrying capacity. Furthermore, platelet count, splenectomy, treatment, and clinical traits were associated with specific omics features, providing insights into GD's clinical heterogeneity and potential diagnostic markers. Autophagy inhibition appears pivotal in GD, driving lipid synthesis, impaired mitochondrial function, and inflammation through chronic activation of mTORC1. Despite limitations like focusing on type 1 GD and using targeted omics approaches, this study provides valuable insights into GD metabolic and immune dysregulation. It lays the basis for future comprehensive investigations into GD manifestations with broader scope and molecular coverage. KEY MESSAGES: The study sheds light on metabolic and immune dysregulation in Gaucher disease. Gaucher disease patients showed elevated phosphatidylcholines, disrupted lipid metabolism, and inflammation profiles. Signs of mitochondrial dysfunction are evident in Gaucher disease patients, with autophagy inhibition significantly affecting lipid synthesis, mitochondrial function, and inflammation via chronic activation of mTORC1.
Collapse
Affiliation(s)
- Franklin Ducatez
- Department of Metabolic Biochemistry, Referral Center for Lysosomal Diseases, Normandie Univ, UNIROUEN, CHU Rouen, INSERM U1245, Filière G2M, 76000, Rouen, France
- Department of Neonatal Pediatrics, Intensive Care and Neuropediatrics, Referral Center for Lysosomal Diseases, Normandie Univ, UNIROUEN, CHU Rouen, INSERM U1245, Filière G2M, 76000, Rouen, France
| | - Marc G Berger
- CHU Clermont-Ferrand, Hopital Estaing, CRB-Auvergne, 63003, Clermont-Ferrand, France
- Université Clermont Auvergne, EA, 7453 CHELTER, 63000, Clermont-Ferrand, France
| | - Carine Pilon
- Department of Metabolic Biochemistry, Referral Center for Lysosomal Diseases, Normandie Univ, UNIROUEN, CHU Rouen, INSERM U1245, Filière G2M, 76000, Rouen, France
| | - Thomas Plichet
- Department of Metabolic Biochemistry, Referral Center for Lysosomal Diseases, Normandie Univ, UNIROUEN, CHU Rouen, INSERM U1245, Filière G2M, 76000, Rouen, France
| | - Céline Lesueur
- Department of Metabolic Biochemistry, Referral Center for Lysosomal Diseases, Normandie Univ, UNIROUEN, CHU Rouen, INSERM U1245, Filière G2M, 76000, Rouen, France
| | - Juliette Berger
- CHU Clermont-Ferrand, Hopital Estaing, CRB-Auvergne, 63003, Clermont-Ferrand, France
- Université Clermont Auvergne, EA, 7453 CHELTER, 63000, Clermont-Ferrand, France
| | - Nadia Belmatoug
- Referral Center for Lysosomal Diseases, Filière G2M, Paris Cité University, APHP-Nord, Paris, France
| | - Stéphane Marret
- Department of Neonatal Pediatrics, Intensive Care and Neuropediatrics, Referral Center for Lysosomal Diseases, Normandie Univ, UNIROUEN, CHU Rouen, INSERM U1245, Filière G2M, 76000, Rouen, France
| | - Soumeya Bekri
- Department of Metabolic Biochemistry, Referral Center for Lysosomal Diseases, Normandie Univ, UNIROUEN, CHU Rouen, INSERM U1245, Filière G2M, 76000, Rouen, France
| | - Abdellah Tebani
- Department of Metabolic Biochemistry, Referral Center for Lysosomal Diseases, Normandie Univ, UNIROUEN, CHU Rouen, INSERM U1245, Filière G2M, 76000, Rouen, France.
| |
Collapse
|
31
|
Shang Q, Zhang X, Pu Y, Lin J, Ma P, Pan Y, Zhao M, Sun D, Cao L. LPCAT1, the Enzyme Responsible for Converting LPC to PC, Promotes OPC Differentiation In Vitro. J Cell Mol Med 2025; 29:e70387. [PMID: 39878319 PMCID: PMC11775935 DOI: 10.1111/jcmm.70387] [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: 04/25/2024] [Revised: 12/06/2024] [Accepted: 01/17/2025] [Indexed: 01/31/2025] Open
Abstract
Myelin is the key structure for high-speed information transmission and is formed by oligodendrocytes (OLs) which are differentiated from oligodendrocyte precursor cells (OPCs) in the central nervous system. Lipid is the main component of myelin and the role of lipid metabolism-related molecules in myelination attach increasing attention. Lysophosphatidylcholine acyltransferase 1 (LPCAT1) mediates the conversion of lysophosphatidylcholine (LPC) to phosphatidylcholine (PC), and its role in myelination draws our interest as LPC is a classical demyelination inducer and PC is a major component of myelin. In this work, LPCAT1 is found expressed in the oligodendrocyte lineage cells during myelination. In vitro experiments showed that the expression level of LPCAT1 gradually increased along with the differentiation process from OPCs to OLs, and over-expression and interference experiments showed that LPCAT1 promoted OPCs differentiation without affecting their proliferation or apoptosis. Mechanistically, the undertaker of LPCAT1's pro-differentiation role is not PC, but the phosphorylated mTOR which is a key regulator in OPCs differentiation. RNA sequencing analysis showed LPCAT1 promoted the expression of ZBTB20 which is an important transcription factor related to lipid metabolism and regulates mTOR phosphorylation. In vivo, complex myelin tomacula involving multiple axons was formed after conditionally knocking out LPCAT1 in oligodendrocyte lineage cells, but no obvious myelin thickness abnormalities were observed. Our results indicate that LPCAT1 is an important regulator of myelination, and lipid metabolism-related molecules may be new valuable targets for the treatment of diseases with myelin abnormalities.
Collapse
Affiliation(s)
- Qi Shang
- Department of Neurobiology, Key Laboratory of Molecular Neurobiology of the Ministry of EducationNaval Medical UniversityShanghaiChina
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain DisordersCapital Medical UniversityBeijingChina
| | - Xin Zhang
- Department of Neurobiology, Key Laboratory of Molecular Neurobiology of the Ministry of EducationNaval Medical UniversityShanghaiChina
| | - Yingyan Pu
- Department of Neurobiology, Key Laboratory of Molecular Neurobiology of the Ministry of EducationNaval Medical UniversityShanghaiChina
| | - Junjian Lin
- Department of Neurobiology, Key Laboratory of Molecular Neurobiology of the Ministry of EducationNaval Medical UniversityShanghaiChina
| | - Peng Ma
- Department of Anesthesiology, Changzheng HospitalNaval Medical UniversityShanghaiChina
| | - Yuchen Pan
- Department of Neurobiology, Key Laboratory of Molecular Neurobiology of the Ministry of EducationNaval Medical UniversityShanghaiChina
| | - Ming Zhao
- Department of Neurology, Naval Specialty Medical CenterNaval Medical UniversityShanghaiChina
| | - Dingya Sun
- Department of Neurobiology, Key Laboratory of Molecular Neurobiology of the Ministry of EducationNaval Medical UniversityShanghaiChina
| | - Li Cao
- Department of Neurobiology, Key Laboratory of Molecular Neurobiology of the Ministry of EducationNaval Medical UniversityShanghaiChina
| |
Collapse
|
32
|
Zhang M, Tang M, Yan K, Zhang Y, Li Y, Tang J, Xu S, Yan X, Hu J, Wang Y. Effects of supplemental medusa (Rhopilema esculentum) on intestinal microbiota and metabolites in silver pomfret (Pampus argenteus). JOURNAL OF FISH BIOLOGY 2025; 106:216-229. [PMID: 39360517 DOI: 10.1111/jfb.15926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 07/23/2024] [Accepted: 08/19/2024] [Indexed: 10/04/2024]
Abstract
Pampus argenteus demonstrates a preference for Rhopilema esculentum as prey, yet the ramifications of consuming supplemental medusa on fish microbiota and metabolism remain elusive. To elucidate these effects, 300 juvenile fish were divided into two groups: control group (C, given commercial food only) and supplemental medusa (SM) group (given supplemental medusa + commercial feed). After 15 days, fish in the SM group exhibited a significant increase in fatness, the amylase activity in the intestine significantly increased, and the intestinal microvilli were arranged more neatly. The comprehensive approach involving 16S rRNA amplicon sequencing and metabolomics was employed, leading to the identification of five genera within the SM group, namely Lactococcus, Cohaesibacter, Maritalea, Sulfitobacter, and Carnobacterium. Functional prediction analysis of the microbiota indicated that the consumption of supplemental medusa facilitated processes such as glycolysis/gluconeogenesis and amino acid absorption. Metabolomics analysis revealed significant enrichment of 85 differential metabolites, most of them belonging to fatty acids and conjugates. These differential metabolites primarily participated in processes such as amino acid metabolism, fatty acid synthesis, and disease. Notably, the consumption of medusa resulted in a significant reduction in nine lysophospholipids associated with cardiovascular disease and inflammation. Pearson's correlation coefficient analysis revealed associations between specific microorganisms and metabolites, indicating that Cobetia, Weissella, and Macrococcus exhibited an increased abundance in the SM group, positively correlating with apocynin, 12-Hete, and delta 9-THC-d3. The indicator bacteria Psychrobacter reduced in the SM group, exhibiting a negative correlation with cystathionine (a compound involved in glutathione synthesis). Overall, the supplementation of medusa may confer a beneficial effect on the immunity of the fish. This study contributes to the theoretical framework for fish feed development.
Collapse
Affiliation(s)
- Man Zhang
- Key Laboratory of Applied Marine Biotechnology, Ningbo University, Ministry of Education, Ningbo, China
- Key Laboratory of Marine Biotechnology of Zhejiang Province, Ningbo University, Ningbo, China
- College of Marine Sciences, Ningbo University, Ningbo, China
| | - Mengke Tang
- Key Laboratory of Applied Marine Biotechnology, Ningbo University, Ministry of Education, Ningbo, China
- Key Laboratory of Marine Biotechnology of Zhejiang Province, Ningbo University, Ningbo, China
- College of Marine Sciences, Ningbo University, Ningbo, China
| | - Kaiheng Yan
- Key Laboratory of Applied Marine Biotechnology, Ningbo University, Ministry of Education, Ningbo, China
- Key Laboratory of Marine Biotechnology of Zhejiang Province, Ningbo University, Ningbo, China
- College of Marine Sciences, Ningbo University, Ningbo, China
| | - Youyi Zhang
- Key Laboratory of Applied Marine Biotechnology, Ningbo University, Ministry of Education, Ningbo, China
- Key Laboratory of Marine Biotechnology of Zhejiang Province, Ningbo University, Ningbo, China
- College of Marine Sciences, Ningbo University, Ningbo, China
| | - Yaya Li
- Key Laboratory of Applied Marine Biotechnology, Ningbo University, Ministry of Education, Ningbo, China
- Key Laboratory of Marine Biotechnology of Zhejiang Province, Ningbo University, Ningbo, China
- College of Marine Sciences, Ningbo University, Ningbo, China
| | - Jie Tang
- Key Laboratory of Applied Marine Biotechnology, Ningbo University, Ministry of Education, Ningbo, China
- Key Laboratory of Marine Biotechnology of Zhejiang Province, Ningbo University, Ningbo, China
- College of Marine Sciences, Ningbo University, Ningbo, China
| | - Shanliang Xu
- Key Laboratory of Applied Marine Biotechnology, Ningbo University, Ministry of Education, Ningbo, China
- Key Laboratory of Marine Biotechnology of Zhejiang Province, Ningbo University, Ningbo, China
- College of Marine Sciences, Ningbo University, Ningbo, China
| | - Xiaojun Yan
- Key Laboratory of Applied Marine Biotechnology, Ningbo University, Ministry of Education, Ningbo, China
- Key Laboratory of Marine Biotechnology of Zhejiang Province, Ningbo University, Ningbo, China
- College of Marine Sciences, Ningbo University, Ningbo, China
| | - Jiabao Hu
- Key Laboratory of Applied Marine Biotechnology, Ningbo University, Ministry of Education, Ningbo, China
- Key Laboratory of Marine Biotechnology of Zhejiang Province, Ningbo University, Ningbo, China
- College of Marine Sciences, Ningbo University, Ningbo, China
| | - Yajun Wang
- Key Laboratory of Applied Marine Biotechnology, Ningbo University, Ministry of Education, Ningbo, China
- Key Laboratory of Marine Biotechnology of Zhejiang Province, Ningbo University, Ningbo, China
- College of Marine Sciences, Ningbo University, Ningbo, China
| |
Collapse
|
33
|
Jokesch P, Oskolkova O, Fedorova M, Gesslbauer B, Bochkov V. Contribution of individual phospholipase A 2 enzymes to the cleavage of oxidized phospholipids in human blood plasma. J Lipid Res 2025; 66:100742. [PMID: 39778770 PMCID: PMC11841071 DOI: 10.1016/j.jlr.2025.100742] [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: 08/29/2024] [Revised: 12/16/2024] [Accepted: 01/03/2025] [Indexed: 01/11/2025] Open
Abstract
Phospholipids containing oxidized esterified PUFA residues (OxPLs) are increasingly recognized for multiple biological activities and causative involvement in disease pathogenesis. Pharmacokinetics of these compounds in blood plasma is essentially not studied. Human plasma contains both genuine phospholipases A2 [platelet activating factor acetyl hydrolase (PAF-AH) (also called Lp-PLA2) and secretory phospholipase A2] and multifunctional enzymes capable of removing sn-2 residues in native and oxidized PLs (lecithin-cholesterol acyltransferase, peroxiredoxin-6). The goal of this study was to compare relative activities of different PLA2 enzymes by analyzing cleavage of oxidized 1-palmitoyl-2-arachidonoyl-sn-glycero-phosphatidylcholine (OxPAPC) and oxidized 1-palmitoyl-2-arachidonoyl-sn-glycero-phosphatidylethanolamine (OxPAPE) by diluted plasma in the presence of enzyme inhibitors. We have found that human plasma demonstrated high total PLA2 activity against oxidized PCs and PEs. PAF-AH/Lp-PLA2 played a dominant role in LysoPC and LysoPE production as compared to other enzymes. Molecular species of oxidized 1-palmitoyl-2-arachidonoyl-sn-glycero-phosphatidylcholine and oxidized 1-palmitoyl-2-arachidonoyl-sn-glycero-phosphatidylethanolamine could be divided into three groups according to their degradation rate and sensitivity to PAF-AH/Lp-PLA2 inhibitor darapladib. Oxidatively truncated species were most rapidly metabolized in the presence of plasma; this process was strongly inhibited by darapladib. The rate of degradation of full-length OxPLs depended on the degree of oxygenation. Species containing 1 to 3 oxygen atoms were relatively stable to degradation in plasma, while OxPLs containing > 3 extra oxygens were degraded but at significantly slower rate than truncated species. In contrast to truncated species, degradation of full-length OxPLs with > 3 extra oxygens were only minimally inhibited by darapladib. These data provide further insights into the mechanisms regulating circulating levels of OxPLs and lipid mediators generated by PLA2 cleavage of OxPLs, namely oxylipins and LysoPC.
Collapse
Affiliation(s)
- Philipp Jokesch
- Institute of Pharmaceutical Sciences, Department of Pharmaceutical Chemistry, University of Graz, Graz, Austria
| | - Olga Oskolkova
- Institute of Pharmaceutical Sciences, Department of Pharmaceutical Chemistry, University of Graz, Graz, Austria
| | - Maria Fedorova
- Center of Membrane Biochemistry and Lipid Research, University Hospital Carl Gustav Carus and Faculty of Medicine of TU Dresden, Dresden, Germany
| | - Bernd Gesslbauer
- Institute of Pharmaceutical Sciences, Department of Pharmaceutical Chemistry, University of Graz, Graz, Austria.
| | - Valery Bochkov
- Institute of Pharmaceutical Sciences, Department of Pharmaceutical Chemistry, University of Graz, Graz, Austria; Field of Excellence BioHealth - University of Graz, Graz, Austria.
| |
Collapse
|
34
|
Kuo H, Lin C, Tsai S, Chen C, Lyu R, Chu C, Ro L, Liao M, Chang H, Weng Y, Hwang J. Blood metabolomic profile in patients with type 2 diabetes mellitus with diabetic peripheral neuropathic pain. J Diabetes Investig 2025; 16:246-256. [PMID: 39548809 PMCID: PMC11786186 DOI: 10.1111/jdi.14355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 10/17/2024] [Accepted: 11/01/2024] [Indexed: 11/18/2024] Open
Abstract
AIMS This study aimed to identify metabolic markers for diabetic peripheral neuropathic pain (DPNP) in patients with type 2 diabetes mellitus (T2DM). MATERIALS AND METHODS Blood metabolite levels in the amino acid, biogenic amine, sphingomyelin, phosphatidylcholine (PC), carnitines, and hexose classes were analyzed in nondiabetic control (n = 27), T2DM without DPNP (n = 58), and T2DM with DPNP (n = 29) using liquid chromatography tandem mass spectrometry. Variable importance projection (VIP) evaluation by partial least squares discriminant analysis was performed on clinical parameters and metabolites. RESULTS Sixteen variables with VIP > 1.0 (P < 0.05) were identified across all patient groups, and 5 variables were identified to discriminate between the two T2DM groups. DPNP patients showed elevated fasting blood glucose, glutamate, PC aa C36:1, lysoPC a C18:1, and lysoPC a C18:2, while low-density lipoprotein cholesterol, phenylalanine, and tryptophan were reduced. Glutamate, lysoPC a C18:1, and lysoPC a C18:2 discriminated T2DM with DPNP from those without DPNP with an AUC of 0.671. The AUC was improved to 0.765 when ratios of metabolite pairs were considered. INTERPRETATION Blood metabolites include glutamate, and phospholipid-related metabolites implicated in neuropathic pain may have the potential as biomarkers for DPNP. Further investigation is required to understand the mechanism of action of these altered metabolites in DPNP.
Collapse
Affiliation(s)
- Hung‐Chou Kuo
- Department of Neurology, Linkou Medical CenterChang Gung Memorial HospitalTaoyuan CityTaiwan
- College of MedicineChang Gung UniversityTaoyuan CityTaiwan
| | - Chia‐Ni Lin
- Department of Laboratory MedicineChang Gung Memorial HospitalTaoyuan CityTaiwan
- Department of Medical Biotechnology and Laboratory Science, College of MedicineChang Gung UniversityTaoyuan CityTaiwan
| | - Sung‐Sheng Tsai
- College of MedicineChang Gung UniversityTaoyuan CityTaiwan
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Linkou Medical CenterChang Gung Memorial HospitalTaoyuan CityTaiwan
| | - Chiung‐Mei Chen
- Department of Neurology, Linkou Medical CenterChang Gung Memorial HospitalTaoyuan CityTaiwan
- College of MedicineChang Gung UniversityTaoyuan CityTaiwan
| | - Rong‐Kuo Lyu
- Department of Neurology, Linkou Medical CenterChang Gung Memorial HospitalTaoyuan CityTaiwan
- College of MedicineChang Gung UniversityTaoyuan CityTaiwan
| | - Chun‐Che Chu
- Department of Neurology, Linkou Medical CenterChang Gung Memorial HospitalTaoyuan CityTaiwan
- College of MedicineChang Gung UniversityTaoyuan CityTaiwan
| | - Long‐Sun Ro
- Department of Neurology, Linkou Medical CenterChang Gung Memorial HospitalTaoyuan CityTaiwan
- College of MedicineChang Gung UniversityTaoyuan CityTaiwan
| | - Ming‐Feng Liao
- Department of Neurology, Linkou Medical CenterChang Gung Memorial HospitalTaoyuan CityTaiwan
- College of MedicineChang Gung UniversityTaoyuan CityTaiwan
| | - Hong‐Shiu Chang
- Department of Neurology, Linkou Medical CenterChang Gung Memorial HospitalTaoyuan CityTaiwan
- College of MedicineChang Gung UniversityTaoyuan CityTaiwan
| | - Yi‐Ching Weng
- Department of Neurology, Linkou Medical CenterChang Gung Memorial HospitalTaoyuan CityTaiwan
- College of MedicineChang Gung UniversityTaoyuan CityTaiwan
| | - Jawl‐Shan Hwang
- College of MedicineChang Gung UniversityTaoyuan CityTaiwan
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Linkou Medical CenterChang Gung Memorial HospitalTaoyuan CityTaiwan
| |
Collapse
|
35
|
Kume H, Kazama K, Sato R, Sato Y. Possible Involvement of Lysophospholipids in Severe Asthma as Novel Lipid Mediators. Biomolecules 2025; 15:182. [PMID: 40001485 PMCID: PMC11852450 DOI: 10.3390/biom15020182] [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: 10/08/2024] [Revised: 12/25/2024] [Accepted: 01/02/2025] [Indexed: 02/27/2025] Open
Abstract
In severe asthma, symptoms are unstable despite intensive treatment based on high doses of inhaled corticosteroids and on-demand use of oral corticosteroids. Although, recently, various biological agents related to Th2 cytokines have been added to intensive controller medications for severe asthma, a significant progress has not been observed in the management for symptoms (dyspnea, wheezing and cough). Medical treatment focused on Type 2 inflammation is probably insufficient to maintain good long-term management for severe asthma. Airway eosinophilia and decreased reversibility in forced expiratory volume in 1 second (FEV1) are listed as major predictors for exacerbation-prone asthma. However, it is generally considered that asthma is complex and heterogeneous. It is necessary to establish precision medicine using treatable traits based on a multidimensional approach related to asthma. Since phospholipids generate lysophospholipids and arachidonic acid by phospholipases, lysophospholipids can be associated with the pathogenesis of this disease via action on smooth muscle, endothelium, and epithelium in the airways. Lysophosphatidic acid (LPA), lysophosphatidylcholine (LPC), and sphingosine 1-phosphate (S1P) are increased in bronchoalveolar fluid after allergen challenge. LPA, LPC, and S1P recruit eosinophils to the lungs and cause β2-adrenergic desensitization. LAP and S1P cause contraction and hyperresponsiveness in airway smooth muscle. Moreover, lysophosphatidylserine and S1P are associated with the allergic reaction related to IgE/FcεRI in mast cells. Lysophospholipid action is probably comprised of corticosteroid resistance and is independent of Type 2 inflammation, and may be corelated with oxidative stress. Lysophospholipids may be a novel molecular target in advancing the management and treatment of asthma. This review discusses the clinical relevance of lysophospholipids in asthma.
Collapse
Affiliation(s)
- Hiroaki Kume
- Department of Infectious Diseases and Respiratory Medicine, Fukushima Medical University Aizu Medical Center, 21-2 Maeda, Tanisawa, Kawahigashi, Aizuwakamatsu 969-3492, Japan; (K.K.); (R.S.); (Y.S.)
| | | | | | | |
Collapse
|
36
|
Mohany KM, Gamal Y, Raheem YFA. Association of LPCAT1-rs8352 genetic variant with susceptibility and severity of pediatric bronchial asthma: a case-control study. BMC Pediatr 2025; 25:68. [PMID: 39871194 PMCID: PMC11770991 DOI: 10.1186/s12887-025-05425-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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 01/13/2025] [Indexed: 01/29/2025] Open
Abstract
BACKGROUND This study aimed to investigate the possible association of LPCAT1-rs8352 genetic variant (single nucleotide change C to G) with the onset and severity of pediatric asthma. Additionally, the study examined the influence of LPCAT1-rs8352 genotypes on asthma-related biomarkers including blood eosinophils count (BEC), eosinophil cationic protein (ECP), high-sensitivity C-reactive protein (hs-CRP), and immunoglobulin E (IgE) and on lung function [forced expiratory volume in one second (FEV1) and the forced vital capacity (FVC)]. PATIENTS AND METHODS The study included ninety-six participant grouped into two groups: G1 (46 asthmatics) and G2 (50 healthy controls). ECP, hs-CRP, and total IgE serum levels were measured using their corresponding ELISA kits. Neonatal blood DNA was extracted using the Gene JET™ Whole Blood Genomic DNA Purification Mini Kit. Genotyping was performed by RT-PCR. RESULTS A significantly higher proportion of individuals in G1 had the LPCAT1-rs8352 CC and GC genotypes compared to G2 (p < 0.001). Individuals with the CC genotype exhibited significantly more severe asthma, along with elevated levels of BEC, ECP, hs-CRP, and total IgE. Those with the GC genotype demonstrated a similar, though less severe, pattern, followed by individuals with the GG genotype. The FEV1 and FVC values showed the opposite trend, with individuals having the GG genotype exhibiting the highest lung function values. CONCLUSION The LPCAT1-rs8352 allele C is associated with pediatric asthma onset and severity. Further research on the LPCAT1 genetic variants may provide a deeper understanding of pediatric bronchial asthma mechanisms and lead to improved management strategies.
Collapse
Affiliation(s)
- Khalid M Mohany
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Assiut University, EL Gammaa street, Assiut, 71515, Egypt.
| | - Yasser Gamal
- Department of Pediatrics, Faculty of Medicine, Assiut University, Egypt, 71515, Assiut, Egypt
| | - Yaser F Abdel Raheem
- Department of Pediatrics, Faculty of Medicine, Assiut University, Egypt, 71515, Assiut, Egypt
| |
Collapse
|
37
|
Arora P, Sharma A, Trivedi R, Sharma P, Padhy S, Shah S, Dutta SK, Manda K, Rana P. Lipidomic Analysis Reveals Systemic Alterations in Servicemen Exposed to Repeated Occupational Low-Level Blast Waves. Mil Med 2025; 190:107-115. [PMID: 38776149 DOI: 10.1093/milmed/usae268] [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: 02/27/2024] [Revised: 04/02/2024] [Accepted: 05/06/2024] [Indexed: 05/24/2024] Open
Abstract
INTRODUCTION Occupational exposure to blast is a prevalent risk experienced by military personnel. While low-level exposure may not manifest immediate signs of illness, prolonged and repetitive exposure may result in neurophysiological dysfunction. Such repeated exposure to occupational blasts has been linked to structural and functional modifications in the brain, adversely affecting the performance of servicemen in the field. These neurological changes can give rise to symptoms resembling concussion and contribute to the development of post-traumatic stress disorder. MATERIALS AND METHODS To understand long-term effects of blast exposure, the study was conducted to assess memory function, serum circulatory protein and lipid biomarkers, and associated concussive symptomology in servicemen. Concussion-like symptoms were assessed using the Rivermead Post-Concussion Symptoms Questionnaire (RPSQ) along with memory function using PGI memory scale. The serum protein biomarkers were quantified using a sandwich ELISA assay, and the serum lipid profile was measured using liquid chromatography-mass spectrometer. RESULTS The findings revealed that repeated low-level blast exposure resulted in impaired memory function, accompanied by elevated levels of serum neurofilament light chain (neuroaxonal injury) and C-reactive protein. Furthermore, alterations in the lipid profile were observed, with an increase in lipid species associated with immune activation. These changes collectively point to systemic inflammation, neuronal injury, and memory dysfunction as pathological characteristics of repeated low-level blast exposure. CONCLUSION The results of our preliminary investigation offer valuable insights for further large-scale study and provide a guiding principle that necessitates a suitable mitigation approach to safeguard the health of personnel against blast overpressure.
Collapse
Affiliation(s)
- Palkin Arora
- Radiological, Nuclear and Imaging Sciences (RNAIS), Institute of Nuclear Medicine and Allied Sciences (INMAS), DRDO, Delhi 110054, India
| | - Apoorva Sharma
- Radiological, Nuclear and Imaging Sciences (RNAIS), Institute of Nuclear Medicine and Allied Sciences (INMAS), DRDO, Delhi 110054, India
| | - Richa Trivedi
- Radiological, Nuclear and Imaging Sciences (RNAIS), Institute of Nuclear Medicine and Allied Sciences (INMAS), DRDO, Delhi 110054, India
| | - Priyanka Sharma
- Radiological, Nuclear and Imaging Sciences (RNAIS), Institute of Nuclear Medicine and Allied Sciences (INMAS), DRDO, Delhi 110054, India
| | - Sankarsan Padhy
- RADAR and Sensor Wing, Proof and Experimental Establishment (PXE), DRDO, Chandipur, Balasore, Odisha 756025, India
| | - Shahnawaj Shah
- RADAR and Sensor Wing, Proof and Experimental Establishment (PXE), DRDO, Chandipur, Balasore, Odisha 756025, India
| | - Suman K Dutta
- Military Wing, Proof and Experimental Establishment (PXE), DRDO, Chandipur, Balasore, Odisha 756025, India
| | - Kailash Manda
- Radiological, Nuclear and Imaging Sciences (RNAIS), Institute of Nuclear Medicine and Allied Sciences (INMAS), DRDO, Delhi 110054, India
| | - Poonam Rana
- Radiological, Nuclear and Imaging Sciences (RNAIS), Institute of Nuclear Medicine and Allied Sciences (INMAS), DRDO, Delhi 110054, India
| |
Collapse
|
38
|
Luettel DM, Terluk MR, Roh J, Weinreb NJ, Kartha RV. Emerging biomarkers in Gaucher disease. Adv Clin Chem 2025; 124:1-56. [PMID: 39818434 DOI: 10.1016/bs.acc.2024.11.001] [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: 01/18/2025]
Abstract
Gaucher disease (GD) is a rare lysosomal disorder characterized by the accumulation of glycosphingolipids in macrophages resulting from glucocerebrosidase (GCase) deficiency. The accumulation of toxic substrates, which causes the hallmark symptoms of GD, is dependent on the extent of enzyme dysfunction. Accordingly, three distinct subtypes have been recognized, with type 1 GD (GD1) as the common and milder form, while types 2 (GD2) and 3 (GD3) are categorized as neuronopathic and severe. Manifestations variably include hepatosplenomegaly, anemia, thrombocytopenia, easy bruising, inflammation, bone pain and other skeletal pathologies, abnormal eye movements and neuropathy. Although the molecular basis of GD is relatively well understood, currently used biomarkers are nonspecific and inadequate for making finer distinctions between subtypes and in evaluating changes in disease status and guiding therapy. Thus, there is continued effort to investigate and identify potential biomarkers to improve GD diagnosis, monitoring and potential identification of novel therapeutic targets. Here, we provide a comprehensive review of emerging biomarkers in GD that can enhance current understanding and improve quality of life through better testing, disease management and treatment.
Collapse
Affiliation(s)
- Danielle M Luettel
- Center for Orphan Drug Research, Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN, United States
| | - Marcia R Terluk
- Center for Orphan Drug Research, Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN, United States
| | - Jaehyeok Roh
- Center for Orphan Drug Research, Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN, United States
| | - Neal J Weinreb
- Department of Human Genetics, Leonard Miller School of Medicine of University of Miami, Miami, FL, United States
| | - Reena V Kartha
- Center for Orphan Drug Research, Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN, United States.
| |
Collapse
|
39
|
Florea VV, Gajjar P, Huang S, Tang J, Zhao S, Davenport M, Mi MY, Haff M, Zhang X, Miller PE, Vasan RS, Liu CT, Lewis GD, Shah RV, Long MT, Nayor M. Hepatic Steatosis and Fibrosis, Cardiorespiratory Fitness, and Metabolic Mediators in the Community. Liver Int 2025; 45:e16147. [PMID: 39673712 DOI: 10.1111/liv.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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 09/25/2024] [Accepted: 10/12/2024] [Indexed: 12/16/2024]
Abstract
BACKGROUND AND AIMS Individuals with steatotic liver disease (SLD) are at high cardiovascular disease (CVD) risk, but approaches to characterise and mitigate this risk are limited. By investigating relations, and shared metabolic pathways, of hepatic steatosis/fibrosis and cardiorespiratory fitness (CRF), we sought to identify new avenues for CVD risk reduction in SLD. METHODS In Framingham Heart Study (FHS) participants (N = 2722, age 54 ± 9 years, 53% women), vibration-controlled transient elastography (VCTE) was performed between 2016-2019 to assess hepatic steatosis (continuous attenuation parameter [CAP]) and fibrosis (liver fibrosis measure [LSM]). Concurrently, participants underwent maximum effort cardiopulmonary exercise testing (CPET), and metabolomic profiling (201 circulating metabolites) was performed in a subsample (N = 1268). RESULTS Mean BMI was 28.0 ± 5.3, 27% had hepatic steatosis, 7.6% had fibrosis, and peak oxygen uptake (VO2) was 26.2 ± 6.8 mL/kg/min in men and 20.7 ± 6.0 mL/kg/min in women (95% predicted overall). In linear models adjusted for cardiometabolic risk factors, greater CAP and LSM were associated with lower peak VO2 (p ≤ 0.002 for all), and the CAP association remained significant after BMI adjustment (p < 0.0001). We observed shared metabolic architecture of CAP, LSM, and peak VO2, with metabolites mediating up to 35% (for CAP) and 74% (for LSM) of the association with peak VO2. Metabolite mediators included amino acids and derivatives implicated in cardiometabolic risk and both protective and deleterious lipid species. CONCLUSIONS Hepatic steatosis and fibrosis are associated with CRF impairment in the community, and these relations are partly mediated by pathways of altered lipid metabolism and general cardiometabolic risk.
Collapse
Affiliation(s)
- Victor V Florea
- Department of Medicine, Mount Auburn Hospital, Harvard Medical School, Cambridge, Massachusetts, USA
| | - Priya Gajjar
- Cardiovascular Medicine Section, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Shi Huang
- Vanderbilt Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Jingxian Tang
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, USA
| | - Shilin Zhao
- Vanderbilt Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Megan Davenport
- Cardiovascular Medicine Section, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Michael Y Mi
- Division of Cardiovascular Medicine, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Madeleine Haff
- Section of Gastroenterology, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Xiaoyu Zhang
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, USA
| | - Patricia E Miller
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, USA
| | - Ramachandran S Vasan
- University of Texas School of Public Health, San Antonio, Texas, USA
- Departments of Medicine and Population Health Sciences, University of Texas Health Science Center, San Antonio, Texas, USA
| | - Ching-Ti Liu
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, USA
| | - Gregory D Lewis
- Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Ravi V Shah
- Vanderbilt Translational and Clinical Research Center, Cardiology Division, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Michelle T Long
- Section of Gastroenterology, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
- Novo Nordisk A/S, Søborg, Denmark
| | - Matthew Nayor
- Cardiovascular Medicine Section, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
- Preventive Medicine and Epidemiology Section, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| |
Collapse
|
40
|
Liu Y, Wu S, Chen L, Teng X, Shi H, Xue C, Li Z. Metabolic profiles and protein expression responses of Pacific oyster (Crassostrea gigas) to polystyrene microplastic stress. Food Chem 2025; 462:140961. [PMID: 39208724 DOI: 10.1016/j.foodchem.2024.140961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 08/01/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024]
Abstract
The underlying toxicity mechanisms of microplastics on oysters have rarely been explored. To fill this gap, the present study investigated the metabolic profile and protein expression responses of oysters to microplastic stress through metabolomics and biochemical analyses. Oysters were exposed to microplastics for 21 days, and the results indicated that the microplastics induced oxidative stress, with a significant decrease in SOD activity in the 0.1 mg/L exposure group. Metabolomics revealed that exposure to microplastics disturbed many metabolic pathways, such as amino acid metabolism, lipid metabolism, biosynthesis of amino acids, aminoacyl-tRNA biosynthesis, and that different concentrations of microplastics induced diverse metabolomic profiles in oysters. Overall, the current study provides new reference data and insights for assessing food safety and consumer health risks caused by microplastic contamination.
Collapse
Affiliation(s)
- Yu Liu
- School of Food Science and Technology, Hainan University, Hainan 570228, PR China; College of Food Science and Engineering, Ocean University of China, No.1299, San Sha Road, Qingdao, Shandong Province 266003, PR China
| | - Shuai Wu
- College of Food Science and Engineering, Ocean University of China, No.1299, San Sha Road, Qingdao, Shandong Province 266003, PR China
| | - Lipin Chen
- School of Food Science and Technology, Hainan University, Hainan 570228, PR China; College of Food Science and Engineering, Ocean University of China, No.1299, San Sha Road, Qingdao, Shandong Province 266003, PR China.
| | - Xiaoyu Teng
- College of Food Science and Engineering, Ocean University of China, No.1299, San Sha Road, Qingdao, Shandong Province 266003, PR China
| | - Haohao Shi
- School of Food Science and Technology, Hainan University, Hainan 570228, PR China; College of Food Science and Engineering, Ocean University of China, No.1299, San Sha Road, Qingdao, Shandong Province 266003, PR China
| | - Changhu Xue
- College of Food Science and Engineering, Ocean University of China, No.1299, San Sha Road, Qingdao, Shandong Province 266003, PR China
| | - Zhaojie Li
- College of Food Science and Engineering, Ocean University of China, No.1299, San Sha Road, Qingdao, Shandong Province 266003, PR China.
| |
Collapse
|
41
|
Zhou Y, Fu K, Li F, Zhang Y, Ren X, Li B, Wu S, Han J, Yang L, Zhou B. UV-aging process of titanium dioxide nanoparticles aggravates enterohepatic toxicity of bis(2-ethylhexyl)-2,3,4,5-tetrabromophthalate to zebrafish. THE SCIENCE OF THE TOTAL ENVIRONMENT 2025; 958:178074. [PMID: 39674164 DOI: 10.1016/j.scitotenv.2024.178074] [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: 09/19/2024] [Revised: 11/12/2024] [Accepted: 12/09/2024] [Indexed: 12/16/2024]
Abstract
The physicochemical characteristics of titanium dioxide nanoparticles (n-TiO2) may change during the aging process once discharged into aquatic environment. However, how the aging process affects their interactions with co-existing pollutants, as well as the joint toxicity has not been explored. This study investigated how UV-aging impacts n-TiO2 in aquatic environments and their interactions with bis(2-ethylhexyl)-2,3,4,5-tetrabromophthalate (TBPH), focusing on their joint toxicity in adult female zebrafish. UV-aging process significantly increased the specific area and hydrophobicity of n-TiO2, promoting the adsorption of TBPH. In vivo experiments revealed that aged n-TiO2 enhanced the bioaccumulation of TBPH in the liver and intestine, worsening hepatic steatosis and intestinal barrier damage. A combined analysis of hepatic lipidomic profiling and intestinal microbiota 16S rRNA sequencing revealed that co-exposure of aged n-TiO2 and TBPH altered gut microbial composition and abundances, facilitating the circulation of lipopolysaccharides (LPS) through the gut-liver axis. Subsequentially, the elevated LPS level in the liver activated the sphingolipid metabolic pathway, resulting in severer lipid metabolism disorders and hepatotoxicity. This study found that UV-aging increases the hydrophobicity and surface area of n-TiO2, enhancing their interaction with the TBPH, which leads to greater bioaccumulation and hepatoxicity through mechanisms involving changes in gut microbiomes and sphingolipid metabolism.
Collapse
Affiliation(s)
- Yuxi Zhou
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Kaiyu Fu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Fan Li
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yindan Zhang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xinxin Ren
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Bingjie Li
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shengmin Wu
- Nanjing Institute of Environmental Sciences, Ministry of Ecology and Environment, Nanjing 210042, China.
| | - Jian Han
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China.
| | - Lihua Yang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Bingsheng Zhou
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| |
Collapse
|
42
|
Youssef AH, Mansour HH, Shousha WG, Galal SM, Abdo SM. Unprecedented Approach for Using Misoprostol Alongside Low-Dose Gamma Radiation to Alleviate Paraquat-Induced Pulmonary Injury in Rats. Dose Response 2025; 23:15593258251326707. [PMID: 40144808 PMCID: PMC11938468 DOI: 10.1177/15593258251326707] [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: 08/05/2024] [Revised: 12/16/2024] [Accepted: 01/23/2025] [Indexed: 03/28/2025] Open
Abstract
Background Abrupt inflammation and alveolar epithelial membrane damage, which may cause the alveolar membrane's malfunction, are related to acute lung injury (ALI). This could eventually lead to pulmonary fibrosis. While lung injury can happen in many ways, the current study will concentrate on the changes in lung pathology mediated by paraquat (PQ). Paraquat, a widely used herbicide, targets lung toxicity through inflammation and oxidative stress, which significantly contribute to lung damage. Objective The current research was to ascertain whether low-dose gamma radiation (R) and misoprostol (MP) could lessen the lung inflammatory cascade started by PQ injection in rats. Methods The ALI model was induced by I.P. injection of PQ (20 mg/kg once), and then treatment was done by MP and/or R for 14 days, and finally, the biochemical and histological parameters were measured in the lung tissues. Results Our data suggest that PQ can promote ALI through TGF-β/smad, Notch, NF-κB, and ET-1 signaling pathways, resulting in EMT. These suggestions were supported by increased levels of TGF-β, inflammatory cytokines, α-SMA, NF-κB, ET-1, CTGF protein, and LPA, whereas PPAR-γ decreased. The aforementioned results have been confirmed by lung histopathology. Conclusion We suggest that the pulmonary inflammatory cascade was hindered and all the previously described gauges improved with R and/or MP therapy.
Collapse
Affiliation(s)
- Ahmed H. Youssef
- Health Radiation Research Department, National Centre for Radiation Research & Technology, Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Heba H. Mansour
- Health Radiation Research Department, National Centre for Radiation Research & Technology, Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Wafaa Gh. Shousha
- Department of Chemistry, Faculty of Science, Helwan University, Cairo, Egypt
| | - Shereen M. Galal
- Health Radiation Research Department, National Centre for Radiation Research & Technology, Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Sara M. Abdo
- Department of Chemistry, Faculty of Science, Helwan University, Cairo, Egypt
| |
Collapse
|
43
|
You Q, Lin Y, Gong JH, Gui WY, Yan QH, Zou JD, Liu EH, Li CY. Integrating lipidomics, 16S rRNA sequencing, and network pharmacology to explore the mechanism of Qikui granule in treating diabetic kidney disease mice. J Chromatogr B Analyt Technol Biomed Life Sci 2025; 1250:124378. [PMID: 39579742 DOI: 10.1016/j.jchromb.2024.124378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 11/01/2024] [Accepted: 11/09/2024] [Indexed: 11/25/2024]
Abstract
Qikui granule (QKG), a hospital preparation of traditional Chinese medicine, has been widely used for diabetic kidney disease (DKD) in clinical practice. However, its holistic therapeutic effects and the underlying therapeutic mechanisms remain unclear. In the present study, the integrated analysis of network pharmacology, 16S rRNA sequencing, and non-targeted lipidomics was performed to explore the anti-DKD effects of QKG and the underlying mechanisms in db/db mouse DKD model. The results of the network pharmacology analysis identified the PI3K-AKT, EGFR, MAPK, JAK-STAT, FoxO, and AGE-RAGE signaling pathways as the potential molecular mechanisms responsible for the efficacy of QKG. Importantly, these signaling pathways were found to be closely related to lipid metabolism and gut microbiota. The therapeutic effectiveness of QKG against DKD was manifested by reducing body weight, alleviating oxidative stress, improving kidney function indicators, promoting the recovery of renal histopathological damage, and regulating the lipid metabolic profile of serum and kidney in db/db mice. A total of 26 lipid metabolites were identified as potential pharmacological biomarkers (PPBs) of QKG for the treatment of DKD, which were mainly involved in glycerophospholipid metabolism. Meanwhile, QKG could alleviate DKD-induced gut microbiota dysbiosis primarily by enriching Candidatus_Arthromitus, which showed a negative correlation with all 26 lipid PPBs as well as 5 biochemical parameters, including 2 oxidative stress factors and 3 kidney function indices. In conclusion, our findings suggest that QKG may upregulate the gut level of Candidatus_Arthromitus to suppress the abnormal activation of PI3K-AKT related signaling pathway, thereby reducing the levels of PC and LPC in the glycerophospholipid metabolism, to finally ameliorate the progression of DKD in db/db mice.
Collapse
Affiliation(s)
- Qing You
- Department of Clinical Pharmacology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, No. 155 Hanzhong Road, Nanjing 210029, China
| | - Yang Lin
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Jia-Hui Gong
- Department of Clinical Pharmacology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, No. 155 Hanzhong Road, Nanjing 210029, China
| | - Wan-Yu Gui
- Department of Clinical Pharmacology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, No. 155 Hanzhong Road, Nanjing 210029, China
| | - Qian-Hua Yan
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Jian-Dong Zou
- Department of Clinical Pharmacology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, No. 155 Hanzhong Road, Nanjing 210029, China.
| | - E-Hu Liu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China; School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China.
| | - Chang-Yin Li
- Department of Clinical Pharmacology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, No. 155 Hanzhong Road, Nanjing 210029, China.
| |
Collapse
|
44
|
Jiang YC, Che Q, Lu X, Liu M, Ye Y, Cao X, Li X, Zhan Y, Dong X, Cheng Y, O’Neill C. Follicular fluid and plasma lipidome profiling and associations towards embryonic development outcomes during ART treatment. Front Endocrinol (Lausanne) 2024; 15:1464171. [PMID: 39790287 PMCID: PMC11712041 DOI: 10.3389/fendo.2024.1464171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Accepted: 12/06/2024] [Indexed: 01/12/2025] Open
Abstract
Introduction It is well acknowledged that lipids assume a critical role in oocyte maturation and early embryonic metabolism, this study aimed to evaluate the relationship between the lipid composition of plasma and follicular fluid (FF), and the consequences of embryonic development. This study compared the lipidomic profiles of paired plasma and FF samples obtained from sixty-five Chinese women who underwent assisted reproductive technology (ART) treatments. Methods Non-targeted lipidomics analysis. Result Results not only indicated similarities in lipid composition between these biofluids, but also revealed a number of unique differences. The biomatrix distinction was found to be primarily driven by lipids belonging to the lysophosphatidylcholines (LPC), phosphatidylethanolamines (PE), ether PE, and triglyceride (TG) classes. In addition, specific species from these subclasses were discovered to be correlated with embryo development outcomes during ART. Notably, the composition of the fatty acyl chains appeared to play a crucial role in these associations. Furthermore, thirteen plasma lipid variables were identified, represented by Phosphatidylcholine 18:014:0 and PE P-18:020:1, which correlated with successful blastocyst formation (BF). Discussion The present study demonstrated that FF has a distinctive lipid composition, setting it apart from plasma; and the association observed with embryonic development underscored an important role of lipid composition in the healthy development of oocytes.
Collapse
Affiliation(s)
- Yingxin Celia Jiang
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- Centenary Institute, The University of Sydney, Sydney, NSW, Australia
| | - Qi Che
- Reproductive Medicine Centre, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xinmei Lu
- Reproductive Medicine Centre, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Miao Liu
- Reproductive Medicine Centre, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yao Ye
- Reproductive Medicine Centre, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiang Cao
- Reproductive Medicine Centre, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xushuo Li
- Center for Tumor Diagnosis & Therapy, Jinshan Hospital, Fudan University, Shanghai, China
| | - Yanxia Zhan
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xi Dong
- Reproductive Medicine Centre, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yunfeng Cheng
- Center for Tumor Diagnosis & Therapy, Jinshan Hospital, Fudan University, Shanghai, China
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai, China
- Institute of Clinical Science, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Christopher O’Neill
- Woolcock Institute for Medical Research, University of Technology, Sydney, NSW, Australia
| |
Collapse
|
45
|
Li J, Li Z, Liu Y, Li Y, Wu Y, Manyande A, Li Z, Xiang H. Lysophosphatidylcholine induced by fat transplantation regulates hyperalgesia by affecting the dysfunction of ACC perineuronal nets. iScience 2024; 27:111274. [PMID: 39640595 PMCID: PMC11617398 DOI: 10.1016/j.isci.2024.111274] [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: 03/04/2024] [Revised: 06/12/2024] [Accepted: 10/25/2024] [Indexed: 12/07/2024] Open
Abstract
The pathogenesis of hyperalgesia is complex and can lead to poor clinical treatment. Our study revealed that epididymal white adipose tissue (eWAT) from spared nerve injury (SNI) mice is involved in the occurrence of hyperalgesia after adipose tissue transplantation. We also showed that lysophosphatidylcholine (LPC) is enriched in the eWAT of SNI mice using non-targeted metabolomic analysis and verified that the levels of LPC in plasma and the anterior cingulate cortex (ACC) region increased following eWAT transplantation. Based on the immunohistochemistry results, we observed that LPC in the ACC region activated microglia via the TRPV1/CamkⅡ pathway. Meanwhile, the disruption of perineuronal nets (PNNs) around PV+ neurons in ACC promoted hyperalgesia, and the loss of PNNs and PV+ interneurons might be due to microglial phagocytosis. These findings elucidate the mechanism underlying hyperalgesia from the perspective of lipid metabolite LPC and PNNs and provide potential strategies for the treatment of hyperalgesia.
Collapse
Affiliation(s)
- Juan Li
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zhen Li
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yanbo Liu
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yijing Li
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yanqiong Wu
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Anne Manyande
- School of Human and Social Sciences, University of West London, London, UK
| | - Zhixiao Li
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Hongbing Xiang
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan 430030, China
| |
Collapse
|
46
|
Wang D, Song J, Wang J, Quan R. Serum metabolic alterations in chickens upon infectious bursal disease virus infection. BMC Vet Res 2024; 20:569. [PMID: 39696379 DOI: 10.1186/s12917-024-04402-3] [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: 09/29/2024] [Accepted: 11/22/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Infectious bursal disease virus (IBDV) is a highly contagious immunosuppressive virus of chickens. Chickens acquire infection by the oral route under natural conditions. Although the histological and pathological changes after IBDV infection are well described, the alterations in serum metabolome have not been reported. In this study, SPF chickens were infected with attenuated IBDV (atIBDV) strain LM and very virulent IBDV (vvIBDV) strain LX, respectively. On the seventh day after oral infection, serum samples of experimental chickens were identified using ultra-high performance liquid chromatography-MS/MS (UHPLC-MS/MS). The serum metabolic profiles were analyzed by multivariate statistical methods. KEGG enrichment analysis was performed to evaluate the dysregulated biological pathways. RESULTS We identified 368 significantly altered metabolites in response to both atIBDV and vvIBDV infection. The metabolic disorder of amino acid and lipid was associated with IBDV infection, especially tryptophan, glycerophospholipid, lysine, and tyrosine metabolism. The differential metabolites enriched in the four metabolic pathways were PC(20:4(5Z,8Z,11Z,14Z)/18:0), PE(16:0/18:2(9Z,12Z)), PE(16:0/22:6(4Z,7Z,10Z,13Z,16Z,19Z)), PE(18:0/20:4(5Z,8Z,11Z,14Z)), PE(18:0/20:4(8Z,11Z,14Z,17Z)), PE(18:0/22:6(4Z,7Z,10Z,13Z,16Z,19Z)), PE(20:3(8Z,11Z,14Z)/16:0), PE(22:6(4Z,7Z,10Z,13Z,16Z,19Z)/16:0), PE-NMe(20:5(5Z,8Z,11Z,14Z,17Z)/18:0), PS(20:3(5Z,8Z,11Z)/18:2(9Z,12Z)), 2-aminobenzoic acid, 4-(2-aminophenyl)-2,4-dioxobutanoic acid, N-acetylserotonin, 5-hydroxyindoleacetate, indole-3-acetaldehyde, indole-3-acetate, p-coumaric acid, L-tyrosine, homovanillin, and S-glutaryldihydrolipoamide. CONCLUSION The atIBDV and vvIBDV infection causes metabolic changes in chicken serum. The differential metabolites and dysregulated metabolic pathways reflect the host response to the IBDV infection.
Collapse
Affiliation(s)
- Dan Wang
- Beijing Key Laboratory for Prevention and mock of Infectious Diseases in Livestock and Poultry, Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, No. 9 Shuguang Garden Middle Road Haidian District, Beijing, 100097, China
| | - Jiangwei Song
- Beijing Key Laboratory for Prevention and mock of Infectious Diseases in Livestock and Poultry, Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, No. 9 Shuguang Garden Middle Road Haidian District, Beijing, 100097, China
| | - Jing Wang
- Beijing Key Laboratory for Prevention and mock of Infectious Diseases in Livestock and Poultry, Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, No. 9 Shuguang Garden Middle Road Haidian District, Beijing, 100097, China
| | - Rong Quan
- Beijing Key Laboratory for Prevention and mock of Infectious Diseases in Livestock and Poultry, Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, No. 9 Shuguang Garden Middle Road Haidian District, Beijing, 100097, China.
| |
Collapse
|
47
|
Taskinen JH, Holopainen M, Ruhanen H, van der Stoel M, Käkelä R, Ikonen E, Keskitalo S, Varjosalo M, Olkkonen VM. Functional omics of ORP7 in primary endothelial cells. BMC Biol 2024; 22:292. [PMID: 39695567 DOI: 10.1186/s12915-024-02087-6] [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: 03/20/2024] [Accepted: 12/02/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Many members of the oxysterol-binding protein-related protein (ORP) family have been characterized in detail over the past decades, but the lipid transport and other functions of ORP7 still remain elusive. What is known about ORP7 points toward an endoplasmic reticulum and plasma membrane-localized protein, which also interacts with GABA type A receptor-associated protein like 2 (GABARAPL2) and unlipidated Microtubule-associated proteins 1A/1B light chain 3B (LC3B), suggesting a further autophagosomal/lysosomal association. Functional roles of ORP7 have been suggested in cholesterol efflux, hypercholesterolemia, and macroautophagy. We performed a hypothesis-free multi-omics analysis of chemical ORP7 inhibition utilizing transcriptomics and lipidomics as well as proximity biotinylation interactomics to characterize ORP7 functions in a primary cell type, human umbilical vein endothelial cells (HUVECs). Moreover, assays on angiogenesis, cholesterol efflux, and lipid droplet quantification were conducted. RESULTS Pharmacological inhibition of ORP7 leads to an increase in gene expression related to lipid metabolism and inflammation, while genes associated with cell cycle and cell division were downregulated. Lipidomic analysis revealed increases in ceramides and lysophosphatidylcholines as well as saturated and monounsaturated triacylglycerols. Significant decreases were seen in all cholesteryl ester and in some unsaturated triacylglycerol species, compatible with the detected decrease of mean lipid droplet area. Along with the reduced lipid stores, ATP-binding cassette subfamily G member 1 (ABCG1)-mediated cholesterol efflux and angiogenesis decreased. Interactomics revealed an interaction of ORP7 with AKT1, a central metabolic regulator. CONCLUSIONS The transcriptomics results suggest an increase in prostanoid as well as oxysterol synthesis, which could be related to the observed upregulation of proinflammatory genes. We envision that the defective angiogenesis in HUVECs subjected to ORP7 inhibition could be the result of an unfavorable plasma membrane lipid composition and/or reduced potential for cell division. To conclude, the present study suggests multifaceted functions of ORP7 in lipid homeostasis, angiogenic tube formation, and gene expression of lipid metabolism, inflammation, and cell cycle in primary endothelial cells.
Collapse
Affiliation(s)
- Juuso H Taskinen
- Minerva Foundation Institute for Medical Research, Tukholmankatu 8, 00290, Helsinki, Finland
| | - Minna Holopainen
- Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, Helsinki University Lipidomics Unit (HiLIPID), Helsinki Institute of Life Science (HiLIFE) and Biocenter Finland, University of Helsinki, Viikinkaari 1, PO BOX 65, 00014, Helsinki, Finland
| | - Hanna Ruhanen
- Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, Helsinki University Lipidomics Unit (HiLIPID), Helsinki Institute of Life Science (HiLIFE) and Biocenter Finland, University of Helsinki, Viikinkaari 1, PO BOX 65, 00014, Helsinki, Finland
| | - Miesje van der Stoel
- Minerva Foundation Institute for Medical Research, Tukholmankatu 8, 00290, Helsinki, Finland
- Department of Anatomy and Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Haartmaninkatu 8, 00290, Helsinki, Finland
| | - Reijo Käkelä
- Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, Helsinki University Lipidomics Unit (HiLIPID), Helsinki Institute of Life Science (HiLIFE) and Biocenter Finland, University of Helsinki, Viikinkaari 1, PO BOX 65, 00014, Helsinki, Finland
| | - Elina Ikonen
- Minerva Foundation Institute for Medical Research, Tukholmankatu 8, 00290, Helsinki, Finland
- Department of Anatomy and Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Haartmaninkatu 8, 00290, Helsinki, Finland
| | - Salla Keskitalo
- Proteomics Unit Viikki, Institute of Biotechnology, HiLIFE and Biocenter Finland, University of Helsinki, Viikinkaari 1, 00790, Helsinki, Finland
| | - Markku Varjosalo
- Proteomics Unit Viikki, Institute of Biotechnology, HiLIFE and Biocenter Finland, University of Helsinki, Viikinkaari 1, 00790, Helsinki, Finland
- Systems Biology/Pathology Research Group, iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, Helsinki, Finland
| | - Vesa M Olkkonen
- Minerva Foundation Institute for Medical Research, Tukholmankatu 8, 00290, Helsinki, Finland.
- Department of Anatomy, Faculty of Medicine, University of Helsinki, Haartmaninkatu 8, Helsinki, 00290, Finland.
| |
Collapse
|
48
|
Jiang S, Fan X, Hua J, Liu S, Feng Y, Shao D, Shen Y, Wang Z, Yan X, Wang J. Integrated metabolomics and network pharmacology analysis to reveal the protective effect of Complanatoside A on nonalcoholic fatty liver disease. Eur J Pharmacol 2024; 985:177074. [PMID: 39481627 DOI: 10.1016/j.ejphar.2024.177074] [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: 07/09/2024] [Revised: 10/11/2024] [Accepted: 10/28/2024] [Indexed: 11/02/2024]
Abstract
INTRODUCTION The rising prevalence and severe consequences of nonalcoholic fatty liver disease (NAFLD) have driven the quest for preventive medications. Complanatoside A (CA) is the marked flavonoid of Astragali complanati semen, a traditional Chinese herb that acts on the liver meridian and is widely used to treat liver problems. CA has been proven to have considerable lipid-lowering and liver-protective effects in vitro. However, the efficacy of CA in preventing NAFLD has yet to be shown in vivo. METHODS First, the effectiveness of CA against NAFLD was assessed using a high-fat diet (HFD) mouse model. Second, the CA protective mechanism against NAFLD was investigated using a combined metabolomics and network pharmacology strategy. Differential metabolites were identified by metabolomics-based analyses, and metabolic pathway analysis was accomplished by MetaboAnalyst. Potential therapeutic targets were obtained through network pharmacology. Finally, key targets were identified via compound-target networks and validated by molecular docking and western blotting. RESULTS CA prevented NAFLD mainly by reducing liver lipid accumulation in HFD mice. Metabolomics identified 22 potential biomarkers for CA treatment of NAFLD, primarily involving glycerophospholipid and arachidonic acid metabolism. Fifty-one potential targets were determined by network pharmacology. Co-analysis revealed that albumin, peroxisome proliferator-activated receptor-alpha, retinoid X receptor alpha, interleukin-6, and tumor necrosis factor alpha were key targets. CONCLUSION This experiment revealed that CA has a preventive effect on NAFLD, primarily by regulating the peroxisome proliferator-activated receptor-alpha/retinoid X receptor alpha pathway. Furthermore, it provides evidence supporting the potential use of CA in the long-term prevention of NAFLD.
Collapse
Affiliation(s)
- Sijia Jiang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, 102488, Beijing, China
| | - Xiaoxu Fan
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, 102488, Beijing, China
| | - Jian Hua
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, 102488, Beijing, China
| | - Shuangqiao Liu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, 102488, Beijing, China
| | - Yingtong Feng
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, 102488, Beijing, China
| | - Danyue Shao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, 102488, Beijing, China
| | - Yiwei Shen
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, 102488, Beijing, China
| | - Zhen Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, 102488, Beijing, China
| | - Xuehua Yan
- Institute of Traditional Chinese Medicine, Xinjiang Medical University, 830011, Urumqi, China.
| | - Jingxia Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, 102488, Beijing, China.
| |
Collapse
|
49
|
Barnette D, Inselman AL, Kaldhone P, Lee GS, Davis K, Sarkar S, Malhi P, Fisher JE, Hanig JP, Beger RD, Jones EE. The incorporation of MALDI mass spectrometry imaging in studies to identify markers of toxicity following in utero opioid exposures in mouse fetuses. FRONTIERS IN TOXICOLOGY 2024; 6:1452974. [PMID: 39691158 PMCID: PMC11651024 DOI: 10.3389/ftox.2024.1452974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 11/07/2024] [Indexed: 12/19/2024] Open
Abstract
Introduction In 2015, the FDA released a Drug Safety Communication regarding a possible link between opioid exposure during early pregnancy and an increased risk of fetal neural tube defects (NTDs). At the time, the indications for opioid use during pregnancy were not changed due to incomplete maternal toxicity data and limitations in human and animal studies. To assess these knowledge gaps, largescale animal studies are ongoing; however, state-of-the-art technologies have emerged as promising tools to assess otherwise non-standard endpoints. Matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI MSI) is a dynamic approach capable of generating 2D ion images to visualize the distribution of an analyte of interest across a tissue section. Methods Given the importance of lipid metabolism and neurotransmitters in the developing central nervous system, this study incorporates MALDI MSI to assess lipid distributions across mouse gestational day (GD) 18 fetuses, with and without observable NTDs following maternal exposure on GD 8 to morphine (400 mg/kg BW) or the NTD positive control valproic acid (VPA) (500 mg/kg BW). Results Analysis of whole-body mouse fetuses revealed differential lipid distributions localized mainly in the brain and spinal cord, which included several phosphatidylcholine (PC) species such as PCs 34:1, 34:0, and 36:2 localized to the cortex or hippocampus and lyso PC 16:0 across all brain regions. Overall, differential lipids increased in with maternal morphine and VPA exposure. Neurotransmitter distributions across the brain using FMP-10 derivatizing agent were also assessed, revealing morphine-specific changes. Discussion The observed differential glycerophospholipid distributions in relation to treatment and NTD development in mouse fetuses provide potential targets for further investigation of molecular mechanisms of opioid-related developmental effects. Overall, these findings support the feasibility of incorporating MALDI MSI to assess non-standard endpoints of opioid exposure during gestation.
Collapse
Affiliation(s)
- Dustyn Barnette
- National Center for Toxicological Research (FDA), Division of Systems Biology, Jefferson, AR, United States
| | - Amy L. Inselman
- National Center for Toxicological Research (FDA), Division of Systems Biology, Jefferson, AR, United States
| | - Pravin Kaldhone
- National Center for Toxicological Research (FDA), Division of Systems Biology, Jefferson, AR, United States
| | - Grace S. Lee
- Center for Drug Evaluation and Research (CDER), Office of Testing and Research, Silver Spring, MD, United States
| | - Kelly Davis
- National Center for Toxicological Research (FDA), Toxicologic Pathology Associates, Jefferson, AR, United States
| | - Sumit Sarkar
- National Center for Toxicological Research (FDA), Division of Neurotoxicology, Jefferson, AR, United States
| | - Pritpal Malhi
- National Center for Toxicological Research (FDA), Toxicologic Pathology Associates, Jefferson, AR, United States
| | - J. Edward Fisher
- Center for Drug Evaluation and Research (CDER), Office of Testing and Research, Silver Spring, MD, United States
| | - Joseph P. Hanig
- Center for Drug Evaluation and Research (CDER), Division of Pharmacology Toxicology for Neuroscience, Silver Spring, MD, United States
| | - Richard D. Beger
- National Center for Toxicological Research (FDA), Division of Systems Biology, Jefferson, AR, United States
| | - E. Ellen Jones
- National Center for Toxicological Research (FDA), Division of Systems Biology, Jefferson, AR, United States
| |
Collapse
|
50
|
Liu LW, Tang M, Zhang ZB, Zhou PP, Xue LP, Jia QQ, Zhao LG, Zuo LH, Sun Z. A stepwise integrated strategy to explore quality markers of Qishen Yiqi dripping pills against myocardial ischemia. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156182. [PMID: 39488103 DOI: 10.1016/j.phymed.2024.156182] [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: 06/13/2024] [Revised: 10/21/2024] [Accepted: 10/21/2024] [Indexed: 11/04/2024]
Abstract
BACKGROUND Numerous experiments and clinical practices have demonstrated the effectiveness of Qishen Yiqi dripping pills (QSYQ) on myocardial ischemia (MI). However, the bioactive ingredients and mechanisms remain unclear, leading to huge gaps between quality control and biological effect of QSYQ. Discovering quality markers (Q-markers) based on effective components is crucial for ensuring stable quality and clinical effectiveness of QSYQ. PURPOSE To explore Q-markers of QSYQ against MI by a stepwise strategy integrating serum pharmacochemistry, network pharmacology, metabolomics, quantitative analysis, and cell experiments. METHODS Firstly, liquid/gas chromatography-mass spectrometry was applied to characterize chemical profiles of QSYQ in vitro and in vivo. Based on the serum migrating constituents, a component-target-MI interaction network was constructed. Subsequently, pharmacodynamics and metabolomics were conducted to evaluate cardioprotective effect and potential mechanism of QSYQ. Next, conjoint analysis of network pharmacology and metabolomics was performed to screen candidate Q-markers. Finally, the measurability and bioactivity were validated to justify their usage as Q-markers. RESULTS A total of 97 components were identified in QSYQ, 24 prototypes of which were detected in serum. The "component-target-disease" interaction network was constructed based on serum migrating constituents. Pharmacodynamic results showed that QSYQ effectively improved cardiac function, attenuated inflammatory cell infiltration, alleviated myocardial fibrosis, and reduced the levels of myocardial enzymes and oxidative stress in MI rats. Metabolomics study demonstrated that 59 metabolites were markedly altered in MI rats, 25 of which were significantly reversely regulated by QSYQ. After integrative analysis of network pharmacology and metabolomics, 12 components were selected as candidate Q-markers of QSYQ, and the contents were quantified. These candidate Q-markers displayed synergistic protective effects against H2O2-induced injury in H9c2 cells. Taken together, 12 components with properties of transitivity and traceability, effectiveness, measurability, and compatibility contribution were defined as representative Q-markers of QSYQ, including Astragaloside IV, Ononin, Calycosin, Formononetin, Rosmarinic acid, Cryptotanshinone, Salvianolic acid A, Tanshinol, Ginsenoside Rb1, Ginsenoside Rg1, Nerolidol, and Santalol. CONCLUSION In this study, a novel stepwise integrated strategy was presented for discovering Q-markers related to therapeutic effects of traditional Chinese medicine prescriptions. Twelve comprehensive and representative Q-markers of QSYQ were identified for the first time to improve its quality control.
Collapse
Affiliation(s)
- Li-Wei Liu
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, 450052, PR China; Henan Engineering Research Center of Clinical Mass Spectrometry for Precision Medicine, Zhengzhou, Henan Province, 450052, PR China
| | - Meng Tang
- The First Department of Orthopaedics, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan Province, 450007, PR China
| | - Zhi-Bo Zhang
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, 450052, PR China; Henan Engineering Research Center of Clinical Mass Spectrometry for Precision Medicine, Zhengzhou, Henan Province, 450052, PR China
| | - Pei-Pei Zhou
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, 450052, PR China; Henan Engineering Research Center of Clinical Mass Spectrometry for Precision Medicine, Zhengzhou, Henan Province, 450052, PR China
| | - Lian-Ping Xue
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, 450052, PR China; Henan Engineering Research Center of Clinical Mass Spectrometry for Precision Medicine, Zhengzhou, Henan Province, 450052, PR China
| | - Qing-Quan Jia
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, 450052, PR China; Henan Engineering Research Center of Clinical Mass Spectrometry for Precision Medicine, Zhengzhou, Henan Province, 450052, PR China
| | - Ling-Guo Zhao
- Center for Disease Prevention and Control of Baoan District, Shenzhen, Guangdong Province, 518101, PR China
| | - Li-Hua Zuo
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, 450052, PR China; Henan Engineering Research Center of Clinical Mass Spectrometry for Precision Medicine, Zhengzhou, Henan Province, 450052, PR China.
| | - Zhi Sun
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, 450052, PR China; Henan Engineering Research Center of Clinical Mass Spectrometry for Precision Medicine, Zhengzhou, Henan Province, 450052, PR China.
| |
Collapse
|