1
|
Alammari AH, Isse FA, O'Croinin C, Davies NM, El-Kadi AOS. Effect of Cannabistilbene I in Attenuating Angiotensin II-Induced Cardiac Hypertrophy: Insights into Cytochrome P450s and Arachidonic Acid Metabolites Modulation. Cannabis Cannabinoid Res 2025; 10:277-288. [PMID: 39324890 DOI: 10.1089/can.2024.0148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/27/2024] Open
Abstract
Introduction: This research investigated the impact of Cannabistilbene I on Angiotensin II (Ang II)-induced cardiac hypertrophy and its potential role in cytochrome P450 (CYP) enzymes and arachidonic acid (AA) metabolic pathways. Cardiac hypertrophy, a response to increased stress on the heart, can lead to severe cardiovascular diseases if not managed effectively. CYP enzymes and AA metabolites play critical roles in cardiac function and hypertrophy, making them important targets for therapeutic intervention. Methods: Adult human ventricular cardiomyocyte cell line (AC16) was cultured and treated with Cannabistilbene I in the presence and absence of Ang II. The effects on mRNA expression related to cardiac hypertrophic markers and CYP were analyzed using real-time polymerase chain reaction, while CYP protein levels were measured by Western blot analysis. AA metabolites were quantified using liquid chromatography-tandem mass spectrometry (LC-MS/MS). Results: Results showed that Ang II triggered hypertrophy, as evidenced by the increase in hypertrophic marker expression, and enlarged the cell surface area, effects that were alleviated by Cannabistilbene I. Gene expression analysis indicated that Cannabistilbene I upregulated CYP1A1, leading to increased enzymatic activity, as evidenced by 7-ethoxyresorufin-O-deethylase assay. Furthermore, LC-MS/MS analysis of AA metabolites revealed that Ang II elevated midchain (R/S)-hydroxyeicosatetraenoic acid (HETE) concentrations, which were reduced by Cannabistilbene I. Notably, Cannabistilbene I selectively increased 19(S)-HETE concentration and reversed the Ang II-induced decline in 19(S)-HETE, suggesting a unique protective role. Conclusion: This study provides new insights into the potential of Cannabistilbene I in modulating AA metabolites and reducing Ang II-induced cardiac hypertrophy, revealing a new candidate as a therapeutic agent for cardiac hypertrophy.
Collapse
Affiliation(s)
- Ahmad H Alammari
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Canada
| | - Fadumo Ahmed Isse
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Canada
| | - Conor O'Croinin
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Canada
| | - Neal M Davies
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Canada
| | - Ayman O S El-Kadi
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Canada
| |
Collapse
|
2
|
Bhat M, Saha P, Narasimhan M, Shelar A, Hole A, Murali Krishna C, Govekar R. Analysis of lipids by Raman spectroscopy and mass spectrometry provides a detection tool and mechanistic insight into imatinib resistance in CML-BC. Biochim Biophys Acta Gen Subj 2025; 1869:130771. [PMID: 39938699 DOI: 10.1016/j.bbagen.2025.130771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 02/03/2025] [Accepted: 02/05/2025] [Indexed: 02/14/2025]
Abstract
Resistance to tyrosine kinase inhibitors (TKIs) is a major challenge in the treatment of chronic myeloid leukemia (CML). Established tests based on the known mechanisms of resistance in the initial chronic phase (CP) confirm resistance, reveal the underlying reason and thereby direct treatment modifications. In the terminal phase of blast crisis (BC), however, additional partially identified mechanisms of resistance exist which necessitates developing modalities to detect resistance regardless of the underlying mechanism and concurrent exploration of the resistance mechanism to assist in identification of appropriate drug targets. In this study both the clinical objectives were achieved by analysing lipids in BC cells, sensitive and resistant to TKIs, using the complementary strengths of distinct analytical technologies. Raman spectroscopy, through the spectral signatures with lipids as a significant differentiating component could segregate resistant from sensitive cells in the Principal Component Analysis (PCA) and Principal Component based Linear Discriminant Analysis (PC-LDA). This provided a tool to rapidly detect resistance in CML-BC despite unclear mechanism of TKI resistance. The depth of coverage achievable by mass spectrometry allowed the generation of quantitative differential profile of individual lipids in resistant cells. The alterations were in diverse classes of lipids which are involved in cell signalling and inhibition studies could link these alterations to modulation of phospholipase A2 (PLA2) levels mediated by p38 mitogen activated protein kinase (p38MAPK), which is causally associated with TKI resistance in CML-BC. Together, lipid analysis using the two platforms, contributed to the detection and mechanistic understanding of imatinib resistance in CML-BC.
Collapse
Affiliation(s)
- Manish Bhat
- Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Navi Mumbai 410210, India; Homi Bhabha National Institute, BARC Training School Complex, Mumbai 400094, India
| | - Panchali Saha
- Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Navi Mumbai 410210, India; Homi Bhabha National Institute, BARC Training School Complex, Mumbai 400094, India
| | - Mythreyi Narasimhan
- School of Biological Sciences, Department of Cell and Developmental Biology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Ashutosh Shelar
- Shimadzu Analytical (India) Pvt. Ltd., Mumbai 400 059, India
| | - Arti Hole
- Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Navi Mumbai 410210, India
| | - C Murali Krishna
- Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Navi Mumbai 410210, India; Homi Bhabha National Institute, BARC Training School Complex, Mumbai 400094, India.
| | - Rukmini Govekar
- Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Navi Mumbai 410210, India; Homi Bhabha National Institute, BARC Training School Complex, Mumbai 400094, India.
| |
Collapse
|
3
|
Guo R, Chang Y, Wang D, Sun H, Gu T, Zong Y, Zhou S, Huang Z, Chen L, Tian Y, Xu W, Lu L, Zeng T. Interaction between cecal microbiota and liver genes of laying ducks with different residual feed intake. Anim Microbiome 2025; 7:30. [PMID: 40119394 PMCID: PMC11929276 DOI: 10.1186/s42523-025-00394-z] [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: 11/05/2024] [Accepted: 03/08/2025] [Indexed: 03/24/2025] Open
Abstract
BACKGROUND The gut microbiota exerts a critical influence on energy metabolism homeostasis and productive performance in avian species. Given the diminishing availability of arable land and intensifying competition for finite resources between livestock production and human populations, the agricultural sector faces dual imperatives to enhance productive efficiency while mitigating ecological footprints. Within this paradigm, optimizing nutrient assimilation efficiency in commercial waterfowl operations emerges as a strategic priority. This investigation employs an integrated multi-omics approach framework (metagenomic, metabolomic, and transcriptomic analyses) to elucidate the mechanistic relationships between cecal microbial consortia and feed conversion ratios in Shan Partridge ducks. RESULTS Based on the analysis of metagenome data, a total of 34 phyla, 1033 genera and 3262 species of bacteria were identified by metagenomic sequencing analysis. At the phylum level, 31 phylums had higher mean abundance in the low residual feed intake ( LRFI) group than in the high residual feed intake (HRFI) group. Among them, the expression of microbiome Elusimicrobiota was significantly higher in the LRFI group than in the HRFI group (P < 0.05). And we also found a significant differences in secondary metabolites biosynthesis, transport, and catabolism pathways between the two groups in microbial function (P < 0.05). Based on metabolomic analysis, 17 different metabolites were found. Among them, Lipids and lipid molecules accounted for the highest proportion. Whereas the liver is very closely related to lipid metabolism, we are close to understanding whether an individual's energy utilization efficiency is related to gene expression in the liver. We selected six ducks from each group of six ducks each for liver transcriptome analysis. A total of 322 differential genes were identified in the transcriptome analysis results, and 319 genes were significantly down-regulated. Among them, we found that prostaglandin endoperoxide synthase 2 (PTGS2) might be a key hub gene regulating RFI by co-occurrence network analysis. Interestingly, the differential gene PTGS2 was enriched in the arachidonic acid pathway at the same time as the differential metabolite 15-deoxy-delta12,14-prostaglandin J2 (15d-PGJ2). In addition, the results of the association analysis of differential metabolites with microorganisms also revealed a significant negative correlation between 15d-PGJ2 and Elusimicrobiota. CONCLUSION Based on comprehensive analysis of the research results, we speculate that the Elusimicrobiota may affect the feed utilization efficiency in ducks by regulating the expression of the PTGS2 gene.
Collapse
Affiliation(s)
- Rongbing Guo
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Livestock and Poultry Resources (Poultry) Evaluation and Utilization, Zhejiang Provincial Engineering Research Center for Poultry Breeding Industry and Green Farming Technology, Institute of Animal Science & Veterinary, Ministry of Agriculture and Rural Affairs of China, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310021, China
| | - Yuguang Chang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Livestock and Poultry Resources (Poultry) Evaluation and Utilization, Zhejiang Provincial Engineering Research Center for Poultry Breeding Industry and Green Farming Technology, Institute of Animal Science & Veterinary, Ministry of Agriculture and Rural Affairs of China, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310021, China
| | - Dandan Wang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Livestock and Poultry Resources (Poultry) Evaluation and Utilization, Zhejiang Provincial Engineering Research Center for Poultry Breeding Industry and Green Farming Technology, Institute of Animal Science & Veterinary, Ministry of Agriculture and Rural Affairs of China, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310021, China
| | - Hanxue Sun
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Livestock and Poultry Resources (Poultry) Evaluation and Utilization, Zhejiang Provincial Engineering Research Center for Poultry Breeding Industry and Green Farming Technology, Institute of Animal Science & Veterinary, Ministry of Agriculture and Rural Affairs of China, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310021, China
| | - Tiantian Gu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Livestock and Poultry Resources (Poultry) Evaluation and Utilization, Zhejiang Provincial Engineering Research Center for Poultry Breeding Industry and Green Farming Technology, Institute of Animal Science & Veterinary, Ministry of Agriculture and Rural Affairs of China, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310021, China
| | - Yibo Zong
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Livestock and Poultry Resources (Poultry) Evaluation and Utilization, Zhejiang Provincial Engineering Research Center for Poultry Breeding Industry and Green Farming Technology, Institute of Animal Science & Veterinary, Ministry of Agriculture and Rural Affairs of China, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310021, China
| | - Shiheng Zhou
- Cherry Valley Agricultural Technology Co. Ltd, Zhoukou, 461300, China
| | - Zhizhou Huang
- Cherry Valley Agricultural Technology Co. Ltd, Zhoukou, 461300, China
| | - Li Chen
- Xianghu Laboratory, Hangzhou, 311231, China
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Livestock and Poultry Resources (Poultry) Evaluation and Utilization, Zhejiang Provincial Engineering Research Center for Poultry Breeding Industry and Green Farming Technology, Institute of Animal Science & Veterinary, Ministry of Agriculture and Rural Affairs of China, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310021, China
| | - Yong Tian
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Livestock and Poultry Resources (Poultry) Evaluation and Utilization, Zhejiang Provincial Engineering Research Center for Poultry Breeding Industry and Green Farming Technology, Institute of Animal Science & Veterinary, Ministry of Agriculture and Rural Affairs of China, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310021, China
| | - Wenwu Xu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Livestock and Poultry Resources (Poultry) Evaluation and Utilization, Zhejiang Provincial Engineering Research Center for Poultry Breeding Industry and Green Farming Technology, Institute of Animal Science & Veterinary, Ministry of Agriculture and Rural Affairs of China, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310021, China
| | - Lizhi Lu
- Xianghu Laboratory, Hangzhou, 311231, China.
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Livestock and Poultry Resources (Poultry) Evaluation and Utilization, Zhejiang Provincial Engineering Research Center for Poultry Breeding Industry and Green Farming Technology, Institute of Animal Science & Veterinary, Ministry of Agriculture and Rural Affairs of China, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310021, China.
| | - Tao Zeng
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Livestock and Poultry Resources (Poultry) Evaluation and Utilization, Zhejiang Provincial Engineering Research Center for Poultry Breeding Industry and Green Farming Technology, Institute of Animal Science & Veterinary, Ministry of Agriculture and Rural Affairs of China, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310021, China.
| |
Collapse
|
4
|
Gui J, Lin W, Meng C, Cui Y, Lan W, He J, Azad MAK, Kong X. Network pharmacology and molecular docking reveal the mechanism of Chinese herb ultrafine powder improving meat nutritional value in aged laying hens. Poult Sci 2025; 104:105047. [PMID: 40138971 PMCID: PMC11985111 DOI: 10.1016/j.psj.2025.105047] [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: 12/26/2024] [Revised: 03/09/2025] [Accepted: 03/15/2025] [Indexed: 03/29/2025] Open
Abstract
This study investigated the effects of dietary Chinese herb ultrafine powder (CHUP) supplementation on meat quality, plasma biochemical parameters, and fatty acid and amino acid composition in pectoral muscles of aged laying hens. A total of 576 Xinyang black-feather laying hens (300-d-old) were randomly allocated to eight groups, including the control group (fed a basal diet) and different CHUP groups (details in 'Materials and methods' section). The trial lasted 120 d. The findings showed that L-LF and L-LF-T supplementation increased the contents of polyunsaturated fatty acids and unsaturated fatty acids (P < 0.05), while CHUP supplementation increased (P < 0.05) the total essential amino acid content in pectoral muscles. Network pharmacology analysis predicted that L-LF-T supplementation mainly influenced the PPAR signaling pathway, which is associated with meat quality. These findings suggest that CHUP supplementation can enhance the nutritional value of pectoral muscles, potentially through its association with the PPAR signaling pathway in aged laying hens.
Collapse
Affiliation(s)
- Jue Gui
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutrition Physiology and Metabolic Processes, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan 410125, China; School of Biology and Food Engineering, Fuyang Normal University, Fuyang, Anhui 236037, China; College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100008, China
| | - Wenchao Lin
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutrition Physiology and Metabolic Processes, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan 410125, China; College of Animal Science and Technology, Hunan Agricultural University, Changsha, Hunan 410128, China
| | - Chengwen Meng
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutrition Physiology and Metabolic Processes, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan 410125, China; College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100008, China
| | - Yadong Cui
- School of Biology and Food Engineering, Fuyang Normal University, Fuyang, Anhui 236037, China
| | - Wei Lan
- School of Biology and Food Engineering, Fuyang Normal University, Fuyang, Anhui 236037, China
| | - Jianhua He
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, Hunan 410128, China
| | - M A K Azad
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutrition Physiology and Metabolic Processes, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan 410125, China; College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100008, China.
| | - Xiangfeng Kong
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutrition Physiology and Metabolic Processes, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan 410125, China; School of Biology and Food Engineering, Fuyang Normal University, Fuyang, Anhui 236037, China; College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100008, China; College of Animal Science and Technology, Hunan Agricultural University, Changsha, Hunan 410128, China.
| |
Collapse
|
5
|
Ma Y, Liu Y, Zhuang L, Dai X, Yao L, Yu J, Zhang L. Metabolomics of the effects of Yishenjiangya granules in older adults with hypertension. Front Pharmacol 2025; 16:1491935. [PMID: 40135238 PMCID: PMC11933063 DOI: 10.3389/fphar.2025.1491935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 02/17/2025] [Indexed: 03/27/2025] Open
Abstract
Introduction Older adults are a high-risk group for hypertension, with specific characteristics regarding symptoms and treatment. Yishenjiangya granules (YJG), a traditional Chinese medicinal decoction, are widely used to reduce blood pressure and improve clinical symptoms. This study aimed to use metabolomics to explore the clinical effects and underlying mechanisms of YJG in hypertension in older adults. Methods The study enrolled patients aged ≥65 years, with systolic blood pressure ≥140 mmHg and/or diastolic blood pressure ≥90 mmHg in sitting positions on different days; the control group comprised 30 healthy participants with normal blood pressure and biochemistry indicators. Ultra-performance liquid chromatography coupled with quadrupole time-of-flight mass spectrometry (UPLC-Q-TOF-MS) was used to analyze plasma metabolites in patients with hypertension before and after YJG intervention. Results After YJG treatment, blood pressure decreased significantly; some metabolites showed a trend approaching the control group. UPLC-Q-TOF-MS analysis identified 30 YJG-targeted plasma metabolites in older adult patients with hypertension, including three major metabolic pathways: linoleic acid, arachidonic acid, and glycerophospholipid metabolism. Conclusion This study identified that metabolite changes may underlie the clinical mechanism of YJG in treating older adult patients with hypertension, providing a basis for further treatment of hypertension.
Collapse
Affiliation(s)
- Yongbo Ma
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
- Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yingying Liu
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Li Zhuang
- Shanghai Changning Tianshan Traditional Chinese Medicine Hospital, Jinan, Shandong, China
| | - Xia Dai
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Li Yao
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Jie Yu
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Lei Zhang
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| |
Collapse
|
6
|
Liu Y, Dai L, Zhang F, Liu Y, Li X, Ma W. Efficacy of In Vitro Addition of Low-Dose Arachidonic Acid in Improving the Sperm Motility of Obese Infertile Men With Asthenozoospermia. J Biochem Mol Toxicol 2025; 39:e70165. [PMID: 39987515 DOI: 10.1002/jbt.70165] [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: 09/18/2024] [Revised: 12/13/2024] [Accepted: 01/16/2025] [Indexed: 02/25/2025]
Abstract
This study aimed to investigate the impact of in vitro low-dose arachidonic acid (AA) addition on enhancing sperm motility in obese infertile men with asthenozoospermia. Semen samples were collected from 115 infertile men, categorized into two BMI groups: 18.5-23.9 kg/m2 and ≥ 28 kg/m2, with all subjects demonstrating a sperm concentration of ≥ 15 × 106/mL. These were further divided into four cohorts based on the percentage of sperm progressive motility (PR): control-normal, control-asthenozoospermia, obese-normal, and obese-asthenozoospermia. Normal PR was classified as ≥ 32%, while asthenozoospermia was characterized by PR < 32%. Metabolomic analysis was employed to quantify seminal plasma metabolites, with differential metabolites identified through statistical evaluation. Additionally, semen samples from 10 infertile men-5 with a body mass index (BMI) of 18.5-23.9 kg/m2 and 5 with a BMI of ≥ 28 kg/m2-underwent further scrutiny. Post-initial semen analysis, 1 mL of semen stock was extracted, treated with 100 pg of AA, incubated at 37°C for 1 h, and reanalyzed to determine the impact on sperm motility. Additionally, 16 Sprague Dawley (SD) rats were split into two groups: control and obese. The control group received a standard diet, while the obese group was subjected to a 45% high-fat diet. After 3 months, the rats were euthanized via cervical dislocation, and their prostate and seminal vesicles were collected for metabolite analysis. A comprehensive analysis of 4635 metabolites in seminal plasma revealed that bile acid secretion emerged as the most significant pathway within the organic systems category, accounting for 0.6% of the total metabolites. Meanwhile, metabolic pathways overwhelmingly dominated the metabolism category, with AA metabolism contributing 4.62%. Notably, 29 metabolites were associated with bile acid secretion, yet no significant differences were observed between the PR ≥ 32% and < 32% groups. In contrast, 214 metabolites were linked to AA metabolism, exhibiting a predominantly downregulated trend, with no upregulated metabolites identified. Within the seminal plasma AA metabolic network, indicators showed a positive association with the induced acrosome reaction, seminal plasma Ca2+ levels, PR, and the proportion of grade A sperm (rapid forward motion, speed ≥ 25 μm/s). Additionally, secretory phospholipase A2 (sPLA2), AA, and cyclooxygenase-1 (COX1) levels demonstrated a negative correlation with anthropometric measurement parameters in the Control-SP group, though this correlation did not reach statistical significance, while a positive correlation was evident in the Obesity-SP group. The concentrations of sPLA2, AA, and COX1 within the AA metabolic network exhibited the following trend: Control-SP-N > Obesity-SP-N > Control-SP-A > Obesity-SP-A. In vitro addition of 100 pg AA significantly enhanced the proportion of grade B sperm (slow-moving, speed < 25 μm/s) while reducing grade C sperm (non-forward-moving) in individuals with a BMI of 18.5-23.9 kg/m2 (p < 0.05). In contrast, for those with a BMI ≥ 28 kg/m2, a marked increase in grade A and grade B sperm and a corresponding reduction in grade C sperm was noted (p < 0.05). Human seminal plasma levels of sPLA2, AA, and COX1 were significantly elevated in the Control-SP group compared to the Obesity-SP group (p < 0.05). However, sPLA2, AA, and COX1 levels in the prostate and seminal vesicle of SD rats did not differ significantly between the Control and Obesity groups (p > 0.05). Distinct metabolic profiles in seminal plasma of infertile men, stratified by BMI, exhibit significant impacts on sperm quality. Low-dose AA, under physiological conditions, maintains sperm integrity and augments fertilization potential. In vitro administration of low-dose AA demonstrates superior effectiveness in enhancing sperm parameters, particularly in obese individuals with asthenozoospermia.
Collapse
Affiliation(s)
- Yongjie Liu
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, School of Basic Medical Sciences, Reproductive Medical Center, Yinchuan Maternity and Child Health Care Hospital, Ningxia Medical University, Yinchuan, China
| | - Liang Dai
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, School of Basic Medical Sciences, Reproductive Medical Center, Yinchuan Maternity and Child Health Care Hospital, Ningxia Medical University, Yinchuan, China
| | - Fan Zhang
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, School of Basic Medical Sciences, Reproductive Medical Center, Yinchuan Maternity and Child Health Care Hospital, Ningxia Medical University, Yinchuan, China
| | - Yang Liu
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, School of Basic Medical Sciences, Reproductive Medical Center, Yinchuan Maternity and Child Health Care Hospital, Ningxia Medical University, Yinchuan, China
| | - Xu Li
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, School of Basic Medical Sciences, Reproductive Medical Center, Yinchuan Maternity and Child Health Care Hospital, Ningxia Medical University, Yinchuan, China
| | - Wenzhi Ma
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, School of Basic Medical Sciences, Reproductive Medical Center, Yinchuan Maternity and Child Health Care Hospital, Ningxia Medical University, Yinchuan, China
| |
Collapse
|
7
|
Shen Y, Zhang G, Wei C, Zhao P, Wang Y, Li M, Sun L. Potential role and therapeutic implications of glutathione peroxidase 4 in the treatment of Alzheimer's disease. Neural Regen Res 2025; 20:613-631. [PMID: 38886929 PMCID: PMC11433915 DOI: 10.4103/nrr.nrr-d-23-01343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 11/27/2023] [Accepted: 12/21/2023] [Indexed: 06/20/2024] Open
Abstract
Alzheimer's disease is an age-related neurodegenerative disorder with a complex and incompletely understood pathogenesis. Despite extensive research, a cure for Alzheimer's disease has not yet been found. Oxidative stress mediates excessive oxidative responses, and its involvement in Alzheimer's disease pathogenesis as a primary or secondary pathological event is widely accepted. As a member of the selenium-containing antioxidant enzyme family, glutathione peroxidase 4 reduces esterified phospholipid hydroperoxides to maintain cellular redox homeostasis. With the discovery of ferroptosis, the central role of glutathione peroxidase 4 in anti-lipid peroxidation in several diseases, including Alzheimer's disease, has received widespread attention. Increasing evidence suggests that glutathione peroxidase 4 expression is inhibited in the Alzheimer's disease brain, resulting in oxidative stress, inflammation, ferroptosis, and apoptosis, which are closely associated with pathological damage in Alzheimer's disease. Several therapeutic approaches, such as small molecule drugs, natural plant products, and non-pharmacological treatments, ameliorate pathological damage and cognitive function in Alzheimer's disease by promoting glutathione peroxidase 4 expression and enhancing glutathione peroxidase 4 activity. Therefore, glutathione peroxidase 4 upregulation may be a promising strategy for the treatment of Alzheimer's disease. This review provides an overview of the gene structure, biological functions, and regulatory mechanisms of glutathione peroxidase 4, a discussion on the important role of glutathione peroxidase 4 in pathological events closely related to Alzheimer's disease, and a summary of the advances in small-molecule drugs, natural plant products, and non-pharmacological therapies targeting glutathione peroxidase 4 for the treatment of Alzheimer's disease. Most prior studies on this subject used animal models, and relevant clinical studies are lacking. Future clinical trials are required to validate the therapeutic effects of strategies targeting glutathione peroxidase 4 in the treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- Yanxin Shen
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, Jilin Province, China
- Cognitive Impairment Center, Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, Jilin Province, China
| | - Guimei Zhang
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, Jilin Province, China
- Cognitive Impairment Center, Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, Jilin Province, China
| | - Chunxiao Wei
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, Jilin Province, China
- Cognitive Impairment Center, Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, Jilin Province, China
| | - Panpan Zhao
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, Jilin Province, China
- Cognitive Impairment Center, Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, Jilin Province, China
| | - Yongchun Wang
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, Jilin Province, China
- Cognitive Impairment Center, Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, Jilin Province, China
| | - Mingxi Li
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, Jilin Province, China
- Cognitive Impairment Center, Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, Jilin Province, China
| | - Li Sun
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, Jilin Province, China
- Cognitive Impairment Center, Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, Jilin Province, China
| |
Collapse
|
8
|
Chen X, He Y, Zhou Y, Gong H, Zhang J, Qiu G, Shen Y, Qin W. Modulatory role of exogenous arachidonic acid in periodontitis with type 2 diabetes mellitus mice. BMC Oral Health 2025; 25:264. [PMID: 39972454 PMCID: PMC11841135 DOI: 10.1186/s12903-025-05525-1] [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: 11/02/2024] [Accepted: 01/20/2025] [Indexed: 02/21/2025] Open
Abstract
BACKGROUND Type 2 diabetes mellitus (T2DM) exhibits a bidirectional relationship with periodontitis, wherein each condition influences the progression of the other. Arachidonic acid (AA) exerts an anti-diabetic effect, while showing a protective effect by regulating the inflammatory response independently of its metabolites. However, its impact on periodontitis with T2DM remains poorly understood. METHODS The T2DM mouse model was established by combining a high-sugar and high-fat diet with streptozotocin injection, followed by silk ligation to induce periodontitis. Alterations in diabetes-associated symptoms were evaluated. Micro-computed tomography was used to measure bone-related parameters, including the distance from the cementoenamel junction to the alveolar bone crest, bone volume/total volume and bone mineral density. Targeted metabolomics analysis was conducted to evaluate the impact of exogenous AA on serum metabolite levels of AA in mice with type 2 diabetic periodontitis. 16S rRNA gene sequencing was utilized to analyze the microbial diversity. The activity of osteoclasts, levels of inflammatory factors and gene expression related to osteoclasts were investigated using TRAP staining and real-time quantitative PCR. RESULTS The periodontitis mouse model with T2DM was successfully established. Following two weeks of exogenous AA treatment, a reduction in fasting blood glucose levels was observed in the diabetic periodontitis mice. Exogenous AA alleviated alveolar bone loss in type 2 diabetic periodontitis mice. However, it had no substantial effect on the contents of serum AA-targeted metabolites. Exogenous AA reduced Staphylococcus in subgingival flora of type 2 diabetic periodontitis mice, but had no significant impact on microbial community structure or diversity. Furthermore, it decreased the number of osteoclasts in the alveolar bone of periodontitis with T2DM mice and increased IL-10 mRNA expression in its gingival tissue. CONCLUSION Exogenous AA may alleviate alveolar bone loss in T2DM mice with periodontitis by reducing the number of osteoclasts and increasing the expression of IL-10 mRNA in periodontal tissues, rather than the change of AA-targeted metabolites in serum or the composition and diversity of microorganisms in subgingival plaque. These findings may provide a potential therapeutic approach for the prevention and treatment of periodontitis with T2DM.
Collapse
Affiliation(s)
- Xiaomin Chen
- Department of Periodontology, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Medical University, Guangzhou, China
| | - Yeqing He
- Department of Periodontology, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Medical University, Guangzhou, China
| | - Yuxi Zhou
- Department of Periodontology, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Medical University, Guangzhou, China
| | - Haihuan Gong
- Department of Periodontology, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Medical University, Guangzhou, China
| | - Jiaming Zhang
- Shenzhen Clinical College of Stomatology, School of Stomatology, Southern Medical University, Shenzhen, China
| | - Guopeng Qiu
- Department of Periodontology, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Medical University, Guangzhou, China
| | - Yuqin Shen
- Department of Periodontology, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Medical University, Guangzhou, China.
| | - Wenguang Qin
- Department of Periodontology, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
9
|
van Eekeren LE, Vadaq N, Blaauw MJT, Groenendijk AL, Vos WAJW, Nelwan EJ, Verbon A, Stalenhoef JE, Berrevoets MAH, van Lunzen J, Netea MG, Weijers G, Riksen NP, Rutten JHW, de Mast Q, Tjwa ETTL, Joosten LAB, van der Ven AJAM. Distinct metabolic perturbations link liver steatosis and incident CVD in lean but not obese PWH. BMC Med 2025; 23:78. [PMID: 39934780 DOI: 10.1186/s12916-025-03914-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 01/27/2025] [Indexed: 02/13/2025] Open
Abstract
BACKGROUND Metabolic dysfunction-associated steatotic liver disease (MASLD) is a key risk factor for cardiovascular disease (CVD), potentially driven by shared metabolic mechanisms. Metabolic perturbations associated with MASLD and CVD remain underexplored in people with HIV (PWH). METHODS We used data from the longitudinal multicenter 2000HIV study comprising 1895 virally suppressed PWH, out of which 970 had available liver and carotid artery measurements. Transient elastography with controlled attenuation parameter (CAP) was performed for the assessment of liver steatosis (CAP > 263 dB/m) and fibrosis (LSM ≥ 7.0). Historic and future incident CVD within 2-year follow-up, defined as myocardial infarction, stroke, peripheral arterial disease, and angina pectoris, were extracted from the medical files, while atherosclerotic plaque(s) in the carotid arteries were assessed using ultrasonography. Metabolic perturbations were analyzed using mass spectrometry-based untargeted metabolomics (n = 500 metabolites) and nuclear magnetic resonance spectroscopy for targeted lipids and other metabolites (n = 246 metabolites). RESULTS PWH with liver steatosis were more likely to have arterial plaques (47% vs. 36%; P value = 0.003) and CVD history (11% vs. 6.8%; P value = 0.021) than PWH without liver steatosis. These associations were only significant in lean PWH, in contrast to those with BMI ≥ 25 kg/m2. Metabolic pathways associated with liver steatosis and fibrosis primarily involved lipid and amino acid metabolism, and they were validated by targeted lipoproteomic measurements. Interestingly, metabolomic pathways and lipoproteomic signatures associated with MASLD were mostly distinct from those associated with CVD parameters. However, several metabolic pathways were shared, especially in lean PWH. These include arachidonic acid metabolism and formation of prostaglandin, purine metabolism, cholecalciferol metabolism, and glycine, serine, alanine, and threonine metabolism. CONCLUSION Metabolic disturbances linked to liver steatosis and CVD diverge across BMI categories in PWH. Lean PWH, unlike their overweight/obese counterparts, show common metabolic perturbations between MASLD and CVD, particularly involving arachidonic acid metabolism. This suggests that lean PWH with liver steatosis may face a heightened risk of CVD due to shared metabolic pathways, potentially opening avenues for targeted interventions, such as aspirin therapy, to mitigate this risk.
Collapse
Affiliation(s)
- Louise E van Eekeren
- Department of Internal Medicine, Radboudumc, Nijmegen, 6500 HB, the Netherlands.
- Radboud Center for Infectious Diseases, Radboudumc, Nijmegen, 6500 HB, the Netherlands.
| | - Nadira Vadaq
- Department of Internal Medicine, Radboudumc, Nijmegen, 6500 HB, the Netherlands
- Radboud Center for Infectious Diseases, Radboudumc, Nijmegen, 6500 HB, the Netherlands
- Division of Tropical Medicine and Infectious Disease, Department of Internal Medicine, Cipto Mangunkusumo Hospital, Jakarta, Indonesia
| | - Marc J T Blaauw
- Department of Internal Medicine, Radboudumc, Nijmegen, 6500 HB, the Netherlands
- Radboud Center for Infectious Diseases, Radboudumc, Nijmegen, 6500 HB, the Netherlands
- Department of Internal Medicine, Elisabeth-Tweesteden Hospital, Tilburg, the Netherlands
| | - Albert L Groenendijk
- Department of Medical Microbiology and Infectious Diseases, Erasmus Medical Center, Rotterdam, the Netherlands
| | | | - Erni J Nelwan
- Division of Tropical Medicine and Infectious Disease, Department of Internal Medicine, Cipto Mangunkusumo Hospital, Jakarta, Indonesia
- Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Annelies Verbon
- Department of Medical Microbiology and Infectious Diseases, Erasmus Medical Center, Rotterdam, the Netherlands
| | | | - Marvin A H Berrevoets
- Department of Internal Medicine, Elisabeth-Tweesteden Hospital, Tilburg, the Netherlands
| | - Jan van Lunzen
- Department of Internal Medicine, Radboudumc, Nijmegen, 6500 HB, the Netherlands
| | - Mihai G Netea
- Department of Internal Medicine, Radboudumc, Nijmegen, 6500 HB, the Netherlands
- Radboud Center for Infectious Diseases, Radboudumc, Nijmegen, 6500 HB, the Netherlands
- Department of Metabolism and Immunology, Life and Medical Sciences Institute, University of Bonn, Bonn, Germany
| | - Gert Weijers
- Medical UltraSound Imaging Center (MUSIC), Division of Medical Imaging, Radboudumc, Nijmegen, the Netherlands
| | - Niels P Riksen
- Department of Internal Medicine, Radboudumc, Nijmegen, 6500 HB, the Netherlands
| | - Joost H W Rutten
- Department of Internal Medicine, Radboudumc, Nijmegen, 6500 HB, the Netherlands
| | - Quirijn de Mast
- Department of Internal Medicine, Radboudumc, Nijmegen, 6500 HB, the Netherlands
- Radboud Center for Infectious Diseases, Radboudumc, Nijmegen, 6500 HB, the Netherlands
| | - Eric T T L Tjwa
- Department of Gastroenterology and Hepatology, Radboudumc, Nijmegen, the Netherlands
| | - Leo A B Joosten
- Department of Internal Medicine, Radboudumc, Nijmegen, 6500 HB, the Netherlands
- Department of Medical Genetics, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca-Napoca, Romania
| | - André J A M van der Ven
- Department of Internal Medicine, Radboudumc, Nijmegen, 6500 HB, the Netherlands
- Radboud Center for Infectious Diseases, Radboudumc, Nijmegen, 6500 HB, the Netherlands
| |
Collapse
|
10
|
Helal SA, Gerges SH, Panahi S, Dyck JRB, El-Kadi AOS. Investigating the sexual dimorphism in isoproterenol-induced cardiac hypertrophy in Sprague Dawley rats. Drug Metab Dispos 2025; 53:100035. [PMID: 39891968 DOI: 10.1016/j.dmd.2025.100035] [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: 10/02/2024] [Accepted: 12/23/2024] [Indexed: 02/03/2025] Open
Abstract
Distinct differences between sexes exist in various cardiovascular diseases. Moreover, there is a significant correlation between the pathogenesis of cardiac hypertrophy (CH) and the metabolites of arachidonic acid (AA) mediated by cytochrome P450 (CYP) enzymes. The potential link between these sex differences, the levels and the activity of CYP enzymes, and their AA-mediated metabolites remains to be elucidated. Male and female Sprague Dawley rats were injected with 1 mg/kg isoproterenol for 7 days to induce CH. Echocardiography was performed before and after the induction of CH. The hypertrophic markers and CYP enzyme levels were analyzed at the gene and protein levels using real-time polymerase chain reaction and Western blot, respectively. Heart microsomal proteins were incubated with AA, and the resulting metabolites were quantified using liquid chromatography-tandem mass spectrometry. Both sexes showed a significant degree of CH, albeit to varying extents, as the echocardiograph, heart weight/tibial length, and left ventricular parameters proved. In addition, the β/α-myosin heavy chain was 2-fold higher in male compared with female rats. Albeit the 20-hydroxyeicosatetraenoic acid (20-HETE) metabolite formation showed no increase in both sexes, the mid-chain HETEs (5- and 15-HETE) were higher in male rats, which paralleled the increase in the gene and protein levels of CYP1B1. The formation rate of the epoxyeicosatrienoic acids was almost unchanged in female-treated rats, while it was significantly decreased in male-treated rats. Our results suggest sexual dimorphism in the isoproterenol-induced CH in rats, specifically on the level of CYP enzymes and their AA-mediated metabolites. SIGNIFICANCE STATEMENT: Sexual dimorphism was observed in rats following isoproterenol-induced cardiac hypertrophy, with males showing a stronger hypertrophic response. This was linked to higher CYP1B1 gene and protein expression in males, along with sex-related differences in many cytochrome P450 enzyme activities and their mediated arachidonic acid metabolites. These findings emphasized the need for targeted, sex-specific therapeutic strategies for the management and treatment of cardiac hypertrophy and other cardiovascular disorders.
Collapse
Affiliation(s)
- Sara A Helal
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada; Faculty of Pharmacy, Department of Biochemistry, Tanta University, Tanta, Egypt
| | - Samar H Gerges
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Sareh Panahi
- Faculty of Medicine & Dentistry, Pediatrics Department, University of Alberta, Edmonton, Alberta, Canada
| | - Jason R B Dyck
- Faculty of Medicine & Dentistry, Pediatrics Department, University of Alberta, Edmonton, Alberta, Canada
| | - Ayman O S El-Kadi
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
11
|
Lu W, Aihaiti A, Abudukeranmu P, Liu Y, Gao H. Arachidonic acid metabolism as a novel pathogenic factor in gastrointestinal cancers. Mol Cell Biochem 2025; 480:1225-1239. [PMID: 38963615 DOI: 10.1007/s11010-024-05057-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 06/25/2024] [Indexed: 07/05/2024]
Abstract
Gastrointestinal (GI) cancers are a major global health burden, representing 20% of all cancer diagnoses and 22.5% of global cancer-related deaths. Their aggressive nature and resistance to treatment pose a significant challenge, with late-stage survival rates below 15% at five years. Therefore, there is an urgent need to delve deeper into the mechanisms of gastrointestinal cancer progression and optimize treatment strategies. Increasing evidence highlights the active involvement of abnormal arachidonic acid (AA) metabolism in various cancers. AA is a fatty acid mainly metabolized into diverse bioactive compounds by three enzymes: cyclooxygenase, lipoxygenase, and cytochrome P450 enzymes. Abnormal AA metabolism and altered levels of its metabolites may play a pivotal role in the development of GI cancers. However, the underlying mechanisms remain unclear. This review highlights a unique perspective by focusing on the abnormal metabolism of AA and its involvement in GI cancers. We summarize the latest advancements in understanding AA metabolism in GI cancers, outlining changes in AA levels and their potential role in liver, colorectal, pancreatic, esophageal, gastric, and gallbladder cancers. Moreover, we also explore the potential of targeting abnormal AA metabolism for future therapies, considering the current need to explore AA metabolism in GI cancers and outlining promising avenues for further research. Ultimately, such investigations aim to improve treatment options for patients with GI cancers and pave the way for better cancer management in this area.
Collapse
Affiliation(s)
- Weiqin Lu
- General Surgery, Cancer Center, Department of Vascular Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | | | | | - Yajun Liu
- Aksu First People's Hospital, Xinjiang, China
| | - Huihui Gao
- Cancer Center, Department of Hospital Infection Management and Preventive Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China.
| |
Collapse
|
12
|
Esmaili H, Tajik B, Tuomainen TP, Kurl S, Salonen JT, Virtanen JK. Associations of serum n-6 polyunsaturated fatty acid concentrations with heart rate at rest, during and after exercise in men. Nutr Metab Cardiovasc Dis 2025:103873. [PMID: 39986931 DOI: 10.1016/j.numecd.2025.103873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 01/20/2025] [Accepted: 01/24/2025] [Indexed: 02/24/2025]
Abstract
BACKGROUND AND AIMS N-6 polyunsaturated fatty acids (PUFA), especially linoleic acid (LA), have been inversely associated with cardiovascular disease (CVD). However, potential mechanisms underlying these associations are not completely known. We evaluated the associations of the serum concentrations of total n-6 PUFA, LA, arachidonic acid (AA), gamma-linolenic acid (GLA), and dihomo-gamma-linolenic acid (DGLA), with resting heart rate (HR), maximal HR during exercise and HR recovery after exercise. METHODS AND RESULTS A total of 872 men free of CVD from the Kuopio Ischaemic Heart Disease Risk Factor Study, aged 42-60 years were studied. The participants performed a maximal symptom-limited exercise stress test with an electrically braked bicycle ergometer. Electrocardiogram reported continuously at rest, during the exercise test, and during recovery. Multivariable-adjusted ANCOVA was used to assess the mean values of resting HR, maximal HR and HR recovery in quartiles of serum n-6 PUFA concentrations. After multivariable adjustments, higher serum LA concentration was associated with lower resting heart rate (extreme-quartile difference = -2.61 beats/min; 95%CI -4.66, -0.56; P-trend = 0.01), but not with maximal HR or HR recovery. Higher concentrations of the minor serum n-6 PUFA GLA and DGLA were only associated with higher maximal HR (for GLA extreme-quartile difference = 2.80 beats/min, 95%CI 0.08,5.52; P-trend = 0.03 and for DGLA extreme-quartile difference = 2.80 beats/min, 95%CI 0.01,5.60; P-trend = 0.03) in the fully adjusted model. AA was not associated with HR. CONCLUSION In conclusion, higher serum LA concentration was associated with lower resting HR, while GLA and DGLA were marginally associated with higher maximal exercise HR. No associations were found with AA.
Collapse
Affiliation(s)
- Haleh Esmaili
- University of Eastern Finland, Kuopio Campus, Institute of Public Health and Clinical Nutrition, Kuopio, Finland
| | - Behnam Tajik
- University of Eastern Finland, Kuopio Campus, Institute of Public Health and Clinical Nutrition, Kuopio, Finland
| | - Tomi-Pekka Tuomainen
- University of Eastern Finland, Kuopio Campus, Institute of Public Health and Clinical Nutrition, Kuopio, Finland
| | - Sudhir Kurl
- University of Eastern Finland, Kuopio Campus, Institute of Public Health and Clinical Nutrition, Kuopio, Finland
| | - Jukka T Salonen
- University of Helsinki, The Faculty of Medicine, Department of Public Health, Finland; MAS-Metabolic Analytical Services Oy, Helsinki, Finland
| | - Jyrki K Virtanen
- University of Eastern Finland, Kuopio Campus, Institute of Public Health and Clinical Nutrition, Kuopio, Finland.
| |
Collapse
|
13
|
Tain YL, Hsu CN. Does maternal consumption of nutritive and non-nutritive sweeteners result in offspring hypertension? Front Nutr 2025; 12:1464269. [PMID: 39911806 PMCID: PMC11794092 DOI: 10.3389/fnut.2025.1464269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Accepted: 01/10/2025] [Indexed: 02/07/2025] Open
Abstract
The consumption of nutritive and non-nutritive sweeteners (NNS) has increased significantly in recent decades. The nutritional status of pregnant women plays a crucial role in determining the likelihood of their offspring developing hypertension in adulthood. While NNSs provide a sweet taste without adding to sugar intake, emerging evidence suggests that maternal consumption of not only nutritive sweeteners (such as fructose) but also NNS may lead to adverse outcomes in offspring, including hypertension. This review provides an overview of the latest research connecting maternal intake of sweeteners to the long-term risk of hypertension in offspring. We examine proposed mechanisms underlying the programming of offspring hypertension by sweeteners, encompassing oxidative stress, dysregulated nutrient sensing signals, abnormal renin-angiotensin system, transcriptome changes, and dysbiotic gut microbiota. Additionally, we outline preventive strategies that can help alleviate offspring hypertension programmed by maternal diets high in sweeteners. Recent advancements in understanding the mechanisms through which maternal consumption of nutritive and non-nutritive sweeteners contributes to offspring hypertension offer promise for addressing this widespread health concern at its developmental roots. Nonetheless, further research is needed to educate the public about the safety of sweetener consumption during pregnancy and lactation.
Collapse
Affiliation(s)
- You-Lin Tain
- Division of Pediatric Nephrology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chien-Ning Hsu
- Department of Pharmacy, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
- School of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
14
|
Fan QQ, Zhai BT, Qiao JX, Zhang D, Sun J, Zhang XF, Sun Y, Bai FY, Guo DY. Study on the underlying mechanism of Huachansu Capsule induced cardiotoxicity of normal rat by integrating transcriptomics, metabolomics and network toxicology. JOURNAL OF ETHNOPHARMACOLOGY 2025; 336:118751. [PMID: 39214192 DOI: 10.1016/j.jep.2024.118751] [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: 06/11/2024] [Revised: 07/31/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Huachansu Capsule (HCSc) is a simple enteric-coated capsule refined from the skin of the dried toad, a traditional medicinal herb. It has been used clinically for many years to treat a variety of malignant tumors with remarkable efficacy. To date, a number of main components of HCSc have been reported to be cardiotoxic, but the specific mechanism of cardiotoxicity is still unknown. AIM OF THE STUDY The aim of this study was to elucidate the possible cardiotoxic symptoms caused by high-doses of HCSc and to further reveal the complex mechanisms by which it causes cardiotoxicity. MATERIALS AND METHODS UPLC-Q-Exactive Orbitrap MS and network toxicology were used to identify and predict the potential toxic components, related signaling pathways. Then, we used acute and sub-acute toxicity experiments to reveal the apparent phenomenon of HCSc-induced cardiotoxicity. Finally, we combined transcriptomics and metabolomics to elucidate the potential mechanism of action, and verified the putative mechanism by molecular docking, RT-qPCR, and Western blot. RESULTS We found 8 toad bufadienolides components may be induced cardiac toxicity HCSc main toxic components. Through toxicity experiments, we found that high dose of HCSc could increase a variety of blood routine indexes, five cardiac enzymes, heart failure indexes (BNP), troponin (cTnI and cTnT), heart rate and the degree of heart tissue damage, while low-dose of HCSc had no such changes. In addition, by molecular docking, found that 8 kinds of main toxic components and cAMP, AMPK, IL1β, mTOR all can be a very good combination, especially in the cAMP. Meanwhile, RT-qPCR and Western blot results showed that HCSc could induce cardiotoxicity by regulating a variety of heart-related differential genes and activating the cAMP signaling pathway. CONCLUSIONS In this study, network toxicology, transcriptomics and metabolomics were used to elucidate the complex mechanism of possible cardiotoxicity induced by high-dose HCSc. Animal experiments, molecular docking, Western blot and RT-qPCR experiments were also used to verify the above mechanism. These findings will inform further mechanistic studies and provide theoretical support for its safe clinical application.
Collapse
Affiliation(s)
- Qiang-Qiang Fan
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an 712046, China
| | - Bing-Tao Zhai
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an 712046, China
| | - Jia-Xin Qiao
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an 712046, China
| | - Dan Zhang
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an 712046, China
| | - Jing Sun
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an 712046, China
| | - Xiao-Fei Zhang
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an 712046, China
| | - Ying Sun
- Shaanxi Dongtai Pharmaceutical Co., Ltd, Xianyang 712031, China
| | - Feng-Yun Bai
- Shaanxi Dongtai Pharmaceutical Co., Ltd, Xianyang 712031, China
| | - Dong-Yan Guo
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an 712046, China.
| |
Collapse
|
15
|
Lv M, Mao X, Lu Z, Yang Y, Huang J, Cheng Y, Ye C, He Z, Shu L, Mo D. 6PPD induces cerebrovascular defects by triggering oxidative stress and ferroptosis in zebrafish. THE SCIENCE OF THE TOTAL ENVIRONMENT 2025; 958:178004. [PMID: 39689467 DOI: 10.1016/j.scitotenv.2024.178004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 12/04/2024] [Accepted: 12/06/2024] [Indexed: 12/19/2024]
Abstract
N-(1,3-dimethylbutyl)-N'-phenyl-p-phenylenediamine (6PPD), which is widely used as an antiozonant in rubber tires, has recently got much attention for its acute aquatic toxicity. However, the developmental toxicity of 6PPD in cerebrovascular network remains unknown. Here, we investigated the effects of 6PPD exposure in cerebral vascular using zebrafish. 6PPD would not affect the body length and shape of zebrafish larvae at the concentrations ranging from 20 μg/L to 1000 μg/L. 6PPD induced developmental defects in the brain in a concentration-dependent manner. The trunk vascular development would not be affected while the cerebrovascular network was disrupted upon 6PPD exposure. 6PPD would trigger excessive Reactive Oxygen Species (ROS) in the brain, indicating abnormal oxidative stress. Mechanistically, brain-specific transcriptome analysis showed that 6PPD could potentially cause the blockage of arachidonic acid (AA) metabolism-related genes and the upregulation of ferroptosis-related genes. Besides, treatment with ferroptosis inhibitor N-Acetyl-L-cysteine (NAC) attenuated oxidative damage and improved the construction of cerebrovascular network upon 6PPD exposure. Moreover, using a human vascular endothelial cell line, we further confirmed that 6PPD could trigger abnormal oxidative stress and defective expansion capacity, implying the conserved toxicity cross species. These findings are useful for the elucidation of toxicity underlying 6PPD in cerebrovascular systems of both zebrafish and humans.
Collapse
Affiliation(s)
- Mengzhu Lv
- Department of Immunology, School of Basic Medicine, Guizhou Medical University, Guiyang 561113, Guizhou, PR China
| | - Xiaoyu Mao
- College of Language Intelligence, Sichuan International Studies University, Chongqing 400031, PR China
| | - Zheng Lu
- Department of Immunology, School of Basic Medicine, Guizhou Medical University, Guiyang 561113, Guizhou, PR China
| | - Yanzhu Yang
- Department of Immunology, School of Basic Medicine, Guizhou Medical University, Guiyang 561113, Guizhou, PR China
| | - Jiangtao Huang
- Department of Immunology, School of Basic Medicine, Guizhou Medical University, Guiyang 561113, Guizhou, PR China
| | - Yuqin Cheng
- College of Animal Science and Technology, Southwest University, Chongqing 400715, PR China
| | - Chuan Ye
- Department of Orthopaedics, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, Guizhou, PR China
| | - Zhixu He
- National & Guizhou Joint Engineering Laboratory for Cell Engineering and Biomedicine Technique, Guizhou Province Key Laboratory for Regenerative Medicine, Guizhou Medical University, Guiyang 561113, Guizhou, PR China
| | - Liping Shu
- Department of Immunology, School of Basic Medicine, Guizhou Medical University, Guiyang 561113, Guizhou, PR China; National & Guizhou Joint Engineering Laboratory for Cell Engineering and Biomedicine Technique, Guizhou Province Key Laboratory for Regenerative Medicine, Guizhou Medical University, Guiyang 561113, Guizhou, PR China.
| | - Dashuang Mo
- Department of Immunology, School of Basic Medicine, Guizhou Medical University, Guiyang 561113, Guizhou, PR China.
| |
Collapse
|
16
|
Poojary G, Vasishta S, Thomas RH, Satyamoorthy K, Padmakumar R, Joshi MB, Babu AS. Exercise improves systemic metabolism in a monocrotaline model of pulmonary hypertension. SPORTS MEDICINE AND HEALTH SCIENCE 2025; 7:37-47. [PMID: 39649790 PMCID: PMC11624410 DOI: 10.1016/j.smhs.2024.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 02/04/2024] [Accepted: 03/04/2024] [Indexed: 12/11/2024] Open
Abstract
Exercise training in pulmonary arterial hypertension (PAH) has been gaining popularity with guidelines now recommending it as an important adjunct to medical therapy. Despite improvements in function and quality of life, an understanding of metabolic changes and their mechanisms remain unexplored. The objective of this study was therefore to understand the metabolic basis of exercise in a monocrotaline model of PAH. 24 male Wistar rats (age: 8-12 weeks and mean body weight: [262.16 ± 24.49] gms) were assigned to one of the four groups (i.e., Control, PAH, Exercise and PAH + Exercise). The exercise groups participated in treadmill running at 13.3 m/min, five days a week for five weeks. Demographic and clinical characteristics were monitored regularly. Following the intervention, LC-MS based metabolomics were performed on blood samples from all groups at the end of five weeks. Metabolite profiling, peak identification, alignment and isotope annotation were also performed. Statistical inference was carried out using dimensionality reducing techniques and analysis of variance. Partial-least-squares discrimination analysis and variable importance in the projection scores showed that the model was reliable, and not over lifting. The analysis demonstrated significant perturbations to lipid and amino acid metabolism, arginine and homocysteine pathways, sphingolipid (p < 0.05), glycerophospholipid (p < 0.05) and nucleotide metabolism in PAH. Exercise, however, was seen to restore arginine (p < 0.05) and homocysteine(p < 0.000 1) levels which were independent effects, irrespective of PAH. Dysregulated arginine and homocysteine pathways are seen in PAH. Exercise restores these dysregulated pathways and could potentially impact severity and outcome in PAH.
Collapse
Affiliation(s)
- Ganesha Poojary
- Department of Physiotherapy, Manipal College of Health Professions, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Sampara Vasishta
- Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - R. Huban Thomas
- Department of Anatomy, Kasturba Medical College Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Kapaettu Satyamoorthy
- SDM College of Medical Sciences and Hospital, Shri Dharmasthala Manjunatheshwara (SDM) University, Manjushree Nagar, Sattur, Dharwad, Karnataka, 580009, India
| | - Ramachandran Padmakumar
- Department of Cardiology, Kasturba Medical College, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Manjunath B. Joshi
- Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Abraham Samuel Babu
- Department of Physiotherapy, Manipal College of Health Professions, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| |
Collapse
|
17
|
Chen W, Yang Y, Zhang Y, Sun C, Ji C, Shen J, Li F, Xiao Y, Wen Y, Liu Q, Zou C. Metabolic profiling reveals altered amino acid and fatty acid metabolism in children with Williams Syndrome. Sci Rep 2024; 14:31467. [PMID: 39733135 PMCID: PMC11682280 DOI: 10.1038/s41598-024-83146-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Accepted: 12/11/2024] [Indexed: 12/30/2024] Open
Abstract
Williams Syndrome (WS) is a rare neurodevelopmental disorder with a prevalence of 1 in 7500 to 1 in 20,000 individuals, caused by a microdeletion in chromosome 7q11.23. Despite its distinctive clinical features, the underlying metabolic alterations remain largely unexplored. This study employs targeted metabolomics to investigate the metabolic characteristics of children with WS. Using liquid chromatography-tandem mass spectrometry (LC-MS/MS), we identified significant dysregulation of 15 metabolites, with 11 upregulated and 4 downregulated. Notably, amino acids such as alanine, proline, and arginine were significantly elevated. Fatty acid metabolism showed pronounced upregulation of long-chain saturated fatty acids (C18:0, C20:0, C22:0, C24:0, C26:0, and C28:0) and downregulation of long-chain unsaturated fatty acids (C18:2 LA, C22:6 DHA, C16:1 PLA, and t-C18:1 EA), except for upregulated nervonic acid (C24:1) and arachidonic acid (C20:4). Metabolic pathway analysis highlighted disruptions in arginine synthesis, arginine/proline metabolism, alanine, aspartate and glutamate metabolism, biosynthesis of unsaturated fatty acids, linoleic acid metabolism, and arachidonic acid metabolism. This study provides the first comprehensive analysis of amino acid and fatty acid metabolism in children with WS, offering insights into the disorder's complex metabolic landscape. Further validation in larger cohorts is essential to confirm these findings and their potential as biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Weijun Chen
- Department of Child Health Care, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, 3333 Binsheng Road, Hangzhou, 310052, Zhejiang Province, China
| | - Yang Yang
- Yunnan Provincial Key Laboratory of Public Health and Biosafety & School of Public Health, Kunming Medical University, Kunming, 650500, People's Republic of China
| | - Yu Zhang
- R&D Department, Zhejiang Biosan Biochemical Technologies Co. Ltd, 859 Shixiang West Rd, Hangzhou, 310007, Zhejiang Province, China
| | - Changxuan Sun
- Suzhou Dushu Lake Hospital, The Fourth Affiliated Hospital of Soochow University, Medical Center of Soochow University, Suzhou, 215123, Jiangshu Province, P. R. China
| | - Chai Ji
- Department of Child Health Care, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, 3333 Binsheng Road, Hangzhou, 310052, Zhejiang Province, China
| | - Jiyang Shen
- Department of Child Health Care, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, 3333 Binsheng Road, Hangzhou, 310052, Zhejiang Province, China
| | - Fangfang Li
- Department of Child Health Care, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, 3333 Binsheng Road, Hangzhou, 310052, Zhejiang Province, China
| | - Yingping Xiao
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Agro-product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Yang Wen
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Agro-product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Qian Liu
- Medical Department, Zhejiang Biosan Biochemical Technologies Co. Ltd, 859 Shixiang West Rd, Hangzhou, 310007, Zhejiang Province, China.
| | - Chaochun Zou
- Department of Endocrinology, Children's Hospital, Zhejiang University School of Medicine, National Children's Regional Medical Center, National Clinical Research Center for Child Health, 3333 Binsheng Road, Hangzhou, 310052, Zhejiang Province, China.
| |
Collapse
|
18
|
Shan S, Jin R, Cheng X, He J, Luo X. Mechano-induced arachidonic acid metabolism promotes keratinocyte proliferation through cPLA2 activity regulation. FASEB J 2024; 38:e70226. [PMID: 39636236 DOI: 10.1096/fj.202402088r] [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: 09/04/2024] [Revised: 11/10/2024] [Accepted: 11/19/2024] [Indexed: 12/07/2024]
Abstract
Mechano-induced keratinocyte hyperproliferation is reported to be associated with various skin diseases. Enhanced cell proliferation often requires the active metabolism of nutrients to produce energy. However, how keratinocytes adapt their cellular metabolism homeostasis to mechanical cues remains unclear. Here, we first found that mechanical stretched keratinocytes showed the accumulation of metabolic arachidonic acid by metabolomic analysis. Second, we found that mechanical stretch promoted keratinocyte proliferation through the activation of cytosolic calcium-dependent phospholipase A2 (cPLA2). Knockdown or inhibition of cPLA2 could reduce the release of arachidonic acid and inhibit the proliferation of stretched keratinocytes in vitro and in vivo. Third, by analyzing overlapping transcriptomes of stretched keratinocytes and arachidonic acid-stimulated keratinocytes, we identified the upregulation of hexokinase domain-containing protein 1 (HKDC1) expression, a novel gene involved in glucose metabolism, which was associated with arachidonic acid-induced keratinocyte proliferation during stretching. Our data reveal a metabolic regulation mechanism by which mechanical stretch induces keratinocyte proliferation, thereby coupling cellular metabolism to the mechanics of the cellular microenvironment. Strategies to change the metabolism process may lead to a new way to treat skin diseases that are related to biophysical forces.
Collapse
Affiliation(s)
- Shengzhou Shan
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rui Jin
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xinwei Cheng
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiahao He
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xusong Luo
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
19
|
Jiang XM, Zhu YL, Gan PR, Li Y, Xia SL, Xu J, Wei Y, Deng R, Wu H. Application of microdialysis combined with lipidomics to analyze fatty acid metabolic changes in the disease process of rheumatoid arthritis. J Pharm Biomed Anal 2024; 251:116448. [PMID: 39216308 DOI: 10.1016/j.jpba.2024.116448] [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: 05/31/2024] [Revised: 08/04/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024]
Abstract
Rheumatoid arthritis (RA) is a metabolically active disease, with shifts in fatty acid metabolism during disease progression profoundly affecting the systemic inflammatory response. Altered fatty acid biomarker metabolism may be a key target for the treatment of RA. To investigate the changes of fatty acid metabolism in RA, collagen-induced arthritis (CIA) model was established. Microdialysis sampling was utilized to overcome the characteristic of occlusive joint cavity in vivo synovial fluid (SF) sampling. Lipidomic methods were established with the UHPLC-Orbitrap Exploris120 platform, and lipid measurements were performed on serum and SF samples. Then, multivariate statistical analyses were performed to detect changes in lipid metabolites induced by CIA. Consequently, a total of 22 potential biomarkers associated with differential fatty acids were screened and identified in serum, and 13 were identified in SF. Notably, alterations were observed in metabolites such as Hexadecanoic acid, Octadecanoic acid, Arachidonic acid, (+/-)11,12-EpETrE, DHA, DPA, Myristic acid, Suberic acid, and others. This study explored a new mechanism of the RA disease process from the perspective of fatty acid metabolism. It provided a new strategy for experimental research on determining the optimal time for establishing CIA model and screening clinical diagnostic biomarkers.
Collapse
Affiliation(s)
- Xiao-Man Jiang
- College of Pharmacy, Anhui University of Chinese Medicine, Qian Jiang Road 1, Hefei 230012, China; Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei 230012, China; Anhui Province Key Laboratory of Research & Development of Chinese Medicine, Hefei 230012, China
| | - Yu-Long Zhu
- College of Pharmacy, Anhui University of Chinese Medicine, Qian Jiang Road 1, Hefei 230012, China; Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei 230012, China; Anhui Province Key Laboratory of Research & Development of Chinese Medicine, Hefei 230012, China
| | - Pei-Rong Gan
- College of Pharmacy, Anhui University of Chinese Medicine, Qian Jiang Road 1, Hefei 230012, China; Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei 230012, China; Anhui Province Key Laboratory of Research & Development of Chinese Medicine, Hefei 230012, China
| | - Ya Li
- College of Pharmacy, Anhui University of Chinese Medicine, Qian Jiang Road 1, Hefei 230012, China; Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei 230012, China; Anhui Province Key Laboratory of Research & Development of Chinese Medicine, Hefei 230012, China
| | - Shi-Lin Xia
- College of Pharmacy, Anhui University of Chinese Medicine, Qian Jiang Road 1, Hefei 230012, China; Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei 230012, China; Anhui Province Key Laboratory of Research & Development of Chinese Medicine, Hefei 230012, China
| | - Jing Xu
- College of Pharmacy, Anhui University of Chinese Medicine, Qian Jiang Road 1, Hefei 230012, China; Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei 230012, China; Anhui Province Key Laboratory of Research & Development of Chinese Medicine, Hefei 230012, China
| | - Yi Wei
- College of Pharmacy, Anhui University of Chinese Medicine, Qian Jiang Road 1, Hefei 230012, China; Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei 230012, China; Anhui Province Key Laboratory of Research & Development of Chinese Medicine, Hefei 230012, China
| | - Ran Deng
- Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei 230012, China; School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, China.
| | - Hong Wu
- College of Pharmacy, Anhui University of Chinese Medicine, Qian Jiang Road 1, Hefei 230012, China; Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei 230012, China; Anhui Province Key Laboratory of Research & Development of Chinese Medicine, Hefei 230012, China.
| |
Collapse
|
20
|
Fan Y, Huang Y, Zhou Y, Ke X, Tian Y, Zheng S, Sun Y, Huang Z, Zhou J, Wu L. Unraveling the liver metabolomic profile of ADB-BUTINACA-induced hepatotoxicity. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 288:117375. [PMID: 39603218 DOI: 10.1016/j.ecoenv.2024.117375] [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: 06/05/2024] [Revised: 11/04/2024] [Accepted: 11/17/2024] [Indexed: 11/29/2024]
Abstract
ADB-BUTINACA, as a new psychoactive substance, can induce physical and psychological dependence. However, the systemic biological impact of ADB-BUTINACA on hepatic metabolomics remains uncertain. The metabolic spectrum in rat livers following exposure to three varying doses of ADB-BUTINACA (0.1, 1, and 5 mg/kg·bw) were analyzed using ultra-high-performance liquid chromatography coupled with high-resolution quadrupole-orbitrap mass spectrometry and molecular docking techniques. Non-target metabolomic technology demonstrated that ADB-BUTINACA induced significant changes in 42 metabolites and disturbed 11 metabolic pathways especially the taurine and hypotaurine metabolism, β-alanine metabolism, and arachidonic acid metabolism, implicates the potential for ADB-BUTINACA to induce not merely cardiac dysfunction but also neurological anomalies. Molecular docking into the hepatotoxic targets of ADB-BUTINACA unveiled its potential for competitive binding with pantetheinase. This interaction may disrupt the coenzyme A (CoA) synthesis pathway, resulting in energy and lipid metabolism imbalances, and ultimately causing hepatotoxic effects. Cellular experiments confirmed reduced HepG2 cell viability and elevated reactive oxygen species (ROS) levels in HepG2 and Huh7 cells. These findings align with our metabolomic findings, supporting the hypothesis that ADB-BUTINACA induces hepatotoxicity via oxidative stress, as well as disruptions in energy and lipid metabolism. This work not only broadens the knowledge of ADB-BUTINACA' toxicological profile but also contributes to efforts aimed at diagnosing and preventing ADB-BUTINACA-induced hepatotoxicity.
Collapse
Affiliation(s)
- Yilei Fan
- Key Laboratory of Drug Prevention and Control Technology of Zhejiang Province, Zhejiang Police College, Hangzhou 310053, China; College of Pharmaceutical Science & Green Pharmaceutical Collaborative Innovation Center of Yangtze River Delta Region, Zhejiang University of Technology, Hangzhou 310053, China
| | - Yingyu Huang
- Department of Food Science and Nutrition, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China
| | - Yi Zhou
- National Narcotics Laboratory Zhejiang Regional Center, Hangzhou 310053, China
| | - Xing Ke
- Key Laboratory of Drug Prevention and Control Technology of Zhejiang Province, Zhejiang Police College, Hangzhou 310053, China
| | - Yimei Tian
- Department of Food Science and Nutrition, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China
| | - Siyue Zheng
- College of Chemical Engineering, Zhejiang University of Technology, Hangzhou 310014, China
| | - Yang Sun
- Hangzhou Bodu Metrology Technology Co., Ltd, Hangzhou 310014, China
| | - Zhongping Huang
- College of Chemical Engineering, Zhejiang University of Technology, Hangzhou 310014, China
| | - Jing Zhou
- Key Laboratory of Drug Prevention and Control Technology of Zhejiang Province, Zhejiang Police College, Hangzhou 310053, China.
| | - Li Wu
- National Narcotics Laboratory Zhejiang Regional Center, Hangzhou 310053, China.
| |
Collapse
|
21
|
Hu R, Xiao J, Fan L. The Role of the Trace Element Selenium in Inflammatory Bowel Disease. Biol Trace Elem Res 2024; 202:4923-4931. [PMID: 38363489 DOI: 10.1007/s12011-024-04074-y] [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: 11/21/2023] [Accepted: 01/17/2024] [Indexed: 02/17/2024]
Abstract
One set of chronic gastrointestinal disorders called inflammatory bowel disease (IBD) is defined by persistent, non-specific inflammation. Abdominal pain, hematochezia, diarrhea, and other symptoms are among its clinical signs. Currently, managing and treating IBD remains a significant challenge. Patients with IBD frequently have deficits in trace elements. Selenium (Se) is one of the necessary trace elements for normal organismal function. It has several regulatory effects, including anti-oxidation, anti-inflammatory, and defensive properties, via inducing the synthesis of selenoproteins. Patients with IBD have been shown to have lower Se levels in epidemiologic research studies. Several experimental models of IBD suggest that Se or selenoproteins play a key role in microinflammation. We discuss the relationship between Se and IBD in this review, with an emphasis on a summary of potential mechanisms of action and applications of Se in IBD.
Collapse
Affiliation(s)
- Ruifang Hu
- College of Clinical Medicine, Jining Medical University, Jining, 272013, Shandong Province, China
| | - Jinliang Xiao
- College of Clinical Medicine, Jining Medical University, Jining, 272013, Shandong Province, China
| | - Lijuan Fan
- Department of Gastroenterology, Jining No. 1 People's Hospital, Jining, 272000, Shandong Province, China.
| |
Collapse
|
22
|
Li Q, Xu X, Zhao C, Wang Y, Chen X, Liu M, Yue C. PUFA and intrahepatic cholestasis of pregnancy: a two-sample Mendelian randomisation analysis. Br J Nutr 2024; 132:1022-1029. [PMID: 39440684 DOI: 10.1017/s0007114524002095] [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: 10/25/2024]
Abstract
This study aimed to explore the potential causal association between PUFA and the risk of intrahepatic cholestasis of pregnancy (ICP) using Mendelian randomisation (MR) analysis. A two-sample MR analysis was conducted utilising large-scale European-based genome-wide association studies summary databases. The primary MR analysis was carried out using the inverse variance-weighted (IVW) method, complemented by other methods such as MR-egger, weighted-median and weighted mode. Sensitivity analysis was also performed to validate the robustness of the findings. Results indicated a 31 % reduced risk of ICP for every 1 standard deviation (sd) increase in n-3 fatty acids levels (OR = 0·69, 95 % CI: 0·54, 0·89, P = 0·004) and in the ratio of n-3 fatty acids to total fatty acids (OR = 0·69, 95 % CI: 0·53, 0·91, P = 0·008). Conversely, there was a 51 % increased risk of ICP for every 1 sd increase in the ratio of n-6 fatty acids to n-3 fatty acids (OR = 1·51, 95 % CI: 1·20, 1·91, P < 0·001) and a 138 % increased risk for every 1 sd increase in the ratio of linoleic fatty acids to total fatty acids (OR = 2·38, 95 % CI: 1·55, 3·66, P < 0·001). The findings suggest that n-3 fatty acids may have a protective effect against the risk of ICP, while n-6 fatty acids and linoleic fatty acids could be potential risk factors for ICP. The supplementation of n-3 fatty acids, as opposed to n-6 fatty acids, could be a promising strategy for the prevention and management of ICP.
Collapse
Affiliation(s)
- Qiong Li
- Department of Obstetrics and Gynecology, The First People's Hospital of Chenzhou, Chenzhou, People's Republic of China
| | - Xinchun Xu
- Department of Ultrasound, Zhangjiagang Hospital Affiliated to Soochow University, Suzhou, People's Republic of China
| | - Chenyang Zhao
- Department of Obstetrics and Gynecology, The First People's Hospital of Chenzhou, Chenzhou, People's Republic of China
| | - Yonghong Wang
- Department of Obstetrics and Gynecology, The First People's Hospital of Chenzhou, Chenzhou, People's Republic of China
| | - Xiaohu Chen
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, People's Republic of China
| | - Miao Liu
- Department of Obstetrics and Gynecology, The First People's Hospital of Chenzhou, Chenzhou, People's Republic of China
| | - Chaoyan Yue
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, People's Republic of China
| |
Collapse
|
23
|
Zhou F, Chang M, Lan Y, Huang W, Sha Z, Liu J, Zhang Z, Ruan S, Liu Z. Effects of saline-alkaline stress on metabolomics profiles, biochemical parameters, and liver histopathology in large yellow croaker (Larimichthys crocea). COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2024; 52:101343. [PMID: 39426067 DOI: 10.1016/j.cbd.2024.101343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 10/10/2024] [Accepted: 10/10/2024] [Indexed: 10/21/2024]
Abstract
China has several saline-alkaline bodies. Studies on the adaptation of fish in saline-alkaline conditions are important for the efficient utilization of such areas. In this study, we employed a comprehensive approach combining histopathological analysis, biochemical markers, and metabolomic profiling to examine the impact of saline-alkaline stress on the liver of the large yellow croaker (Larimichthys crocea). It was found that the survival rate of L. crocea in the saline-alkaline treated group (EX) was significantly higher than that of the control group (CK). Saline-alkaline stress could not influence the structure of the liver of L. crocea, and not change the levels of superoxide dismutase (SOD), catalase (CAT), alkaline phosphatase (ALP), acid phosphatase (ACP). In addition, we identified 5953 metabolites, and 312 differentially expressed metabolites (DEMs) showed significant differential expression between the CK and EX groups. In the positive ion mode, 216 DEMs were identified, including 120 up-regulated and 96 down-regulated DEMs, and in the negative ion mode, 178 DEMs were identified, including 131 up-regulated and 47 down-regulated DEMs. Pathway enrichment analysis revealed significant involvement in 58 metabolic pathways, primarily linked to energy metabolism. These included the metabolism of amino acid, carbohydrate, and lipid pathways, including cysteine and methionine metabolism, biosynthesis of valine, leucine, isoleucine, and ascorbate; aldarate metabolism; galactose metabolism; glycerophospholipid metabolism; and the biosynthesis of unsaturated fatty acids. Metabolomics revealed that increased synthesis of compounds, such as succinic acid, arachidonic acid, and L-gulonic acid in the liver of L.crocea, is associated with adaptation to saline-alkaline aquaculture conditions. The findings of this study indicated that the fish mitigate reactive oxygen species induced by hyperosmotic environments and improve cellular membrane fluidity and intercellular signal transduction through the metabolism of unsaturated fatty acids and carbohydrates, facilitating adaptation to saline-alkaline conditions.
Collapse
Affiliation(s)
- Fengfang Zhou
- College of Biological Science and Engineering, Ningde Normal University, Ningde 352100, China
| | - Mengyang Chang
- Institute of Aquatic Biotechnology, College of Life Sciences, Qingdao University, Qingdao 266071, China
| | - Yan Lan
- Institute of Aquatic Biotechnology, College of Life Sciences, Qingdao University, Qingdao 266071, China
| | - Weiqing Huang
- College of Marine Sciences, Ningde Normal University, Ningde 352100, China.
| | - Zhenxia Sha
- Institute of Aquatic Biotechnology, College of Life Sciences, Qingdao University, Qingdao 266071, China.
| | - Jiafu Liu
- College of Marine Sciences, Ningde Normal University, Ningde 352100, China.
| | - Zipeng Zhang
- Ningxia Lanwan Ecological Agriculture Co., Ltd., Yinchuan 750000, China
| | - Shaojiang Ruan
- College of Biological Science and Engineering, Ningde Normal University, Ningde 352100, China
| | - Zheng Liu
- College of Biological Science and Engineering, Ningde Normal University, Ningde 352100, China
| |
Collapse
|
24
|
Zhang Z, Zheng Y, Zhang B, Wang R, Chen L, Wang Y, Feng W, Zheng X, Li K, Zhou N. Untargeted serum and gastric metabolomics and network pharmacology analysis reveal the superior efficacy of zingiberis rhizoma recens-/euodiae fructus-processed Coptidis Rhizoma on gastric ulcer rats. JOURNAL OF ETHNOPHARMACOLOGY 2024; 332:118376. [PMID: 38782310 DOI: 10.1016/j.jep.2024.118376] [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: 03/19/2024] [Revised: 05/08/2024] [Accepted: 05/20/2024] [Indexed: 05/25/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Zingiberis rhizoma recens-/wine-/euodiae fructus-processed Coptidis Rhizoma (CR, zCR/wCR/eCR) are the commonly used processed products of CR in clinic. After being processed with different excipients, the efficacy of CR will change accordingly. I.e., wCR could resolve excessive heat of the upper energizer, zCR could eliminate gastric heat and harmonize the stomach, eCR could smooth the liver and harmonize the stomach. However, the underlying mechanisms were still unclear. AIM OF THE STUDY To further verify the differential efficacy of the three processed CR products and compare the mechanisms on gastric ulcer. MATERIAL AND METHODS First, a GU model, whose onset is closely related to the heat in stomach and the disharmony between liver and stomach, was established, and the therapeutic effects of zCR/wCR/eCR/CR were evaluated by pathologic observation and measurement of cytokine levels. Second, metabolomics analysis and network pharmacology were conducted to reveal the differential intervening mechanism of zCR/eCR on GU. Third, the predicted mechanisms from metabolomics analysis and network pharmacology were validated using western blotting, flow cytometry and immunofluorescence. RESULTS zCR/wCR/eCR/CR could alleviate the pathologic damage to varying degrees. In metabolomics research, fewer metabolic pathways were enriched in serum samples, and most of them were also present in the results of gastric tissue samples. The gastroprotective, anti-inflammatory, antioxidant, and anti-apoptotic effects of zCR/wCR/eCR/CR might be due to their interference on histidine, arachidonic acid, and glycerophospholipids metabolism. Quantitative results indicated that zCR/eCR had a better therapeutic effect than wCR/CR in treating GU. A comprehensive analysis of metabolomics and network pharmacology revealed that zCR and eCR exerted anti-GU effects via intervening in five core targets, including AKT, TNF, IL6, IL1B and PPARG. In the validation experiment, zCR/eCR could significantly reverse the abnormal expression of proteins related to apoptosis, inflammation, oxidative stress, gastric function, as well as the PI3K/AKT signaling pathways. CONCLUSION zCR and eCR could offer gastroprotective benefits by resisting inflammation and apoptosis, inhibiting gastric-acid secretion, as well as strengthening gastric mucosal defense and antioxidant capacity. Integrating network pharmacology and metabolomics analysis could reveal the acting mechanism of drugs and promote the development of medications to counteract GU.
Collapse
Affiliation(s)
- Zhenkai Zhang
- Henan University of Chinese Medicine, Zhengzhou, 450046, PR China
| | - Yajuan Zheng
- Henan University of Chinese Medicine, Zhengzhou, 450046, PR China
| | - Bingxian Zhang
- Henan University of Chinese Medicine, Zhengzhou, 450046, PR China
| | - Ruifeng Wang
- Henan University of Chinese Medicine, Zhengzhou, 450046, PR China
| | - Long Chen
- Henan University of Chinese Medicine, Zhengzhou, 450046, PR China; Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Zhengzhou, 450046, PR China
| | - Yongxiang Wang
- Henan University of Chinese Medicine, Zhengzhou, 450046, PR China
| | - Weisheng Feng
- Henan University of Chinese Medicine, Zhengzhou, 450046, PR China; Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases By Henan & Education Ministry of PR China, Zhengzhou, 450046, PR China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, 450046, PR China.
| | - Xiaoke Zheng
- Henan University of Chinese Medicine, Zhengzhou, 450046, PR China; Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases By Henan & Education Ministry of PR China, Zhengzhou, 450046, PR China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, 450046, PR China.
| | - Kai Li
- Henan University of Chinese Medicine, Zhengzhou, 450046, PR China; Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Zhengzhou, 450046, PR China.
| | - Ning Zhou
- Henan University of Chinese Medicine, Zhengzhou, 450046, PR China; Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Zhengzhou, 450046, PR China; Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases By Henan & Education Ministry of PR China, Zhengzhou, 450046, PR China.
| |
Collapse
|
25
|
Wang R, Zhang X, Wang Y, Lin Y, Zhou Y, Wang Y, Li G. Gut microbiota of miR-30a-5p-deleted mice aggravate high-fat diet-induced hepatic steatosis by regulating arachidonic acid metabolic pathway. Clin Transl Med 2024; 14:e70035. [PMID: 39360667 PMCID: PMC11447637 DOI: 10.1002/ctm2.70035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 09/08/2024] [Accepted: 09/15/2024] [Indexed: 10/04/2024] Open
Abstract
BACKGROUND Patients with non-alcoholic fatty liver disease (NAFLD) often exhibit hepatic steatosis and dyslipidemia. Studies have shown that intestinal microorganisms are closely related to the occurrence of NAFLD and atherosclerosis. Our previous study has underscored the protective role of microRNA-30a-5p (miR-30a-5p) against atherosclerosis. METHODS AND RESULTS In the present study, we aimed to elucidate the effect and underlying mechanism of the intestinal microorganisms of miR-30a-5p knockout (KO) mice on NAFLD. Our findings demonstrated that KO exacerbated high-fat diet (HFD)-induced hepatic steatosis and disrupted liver function, as evidenced by elevated levels of total cholesterol, low-density lipoprotein, alanine aminotransferase, aspartate transaminase, and total bile acids in serum. Fecal microbiota from HFD-fed KO mice induced hepatic steatosis, dyslipidemia, and higher levels of enzymes indicative of liver damage in wild-type mice. Remarkably, KO mice significantly intensified the above effects. 16s rDNA sequencing and metabolomics of the intestinal microbiota in the HFD-treated KO and WT mice showed that the loss of miR-30a-5p resulted in intestinal microbiota imbalance and was highly related to the arachidonic acid metabolic pathway. Targeted metabolomic in the liver tissues unveiled upregulation of COX-related (PGF2a, 8-iso-PGF2a and PGF2) and LOX-related (LTB4, LTD4, 12S-HETE and 15S-HETE) factors in HFD-treated KO mice. Immunohistochemistry and transcriptional analyses showed that miR-30a-5p affected arachidonic acid metabolism through the LOX/COX pathways. Besides, COX/LOX pathways and hepatic steatosis were reversed after reintroducing miR-30a-5p in HFD-treated KO mice. CONCLUSIONS This study reveals the pivotal mechanism by which miR-30a-5p and intestinal microbes regulate hepatic steatosis and abnormal lipid metabolism, offering promising avenues for NAFLD and atherosclerosis therapeutics. HIGHLIGHTS MiR-30a-5p deletion aggravated hepatic steatosis and lipid disorder induced by an HFD in mice. Gut microbiota participated in the regulation of hepatic steatosis in the context of miR-30a-5p. Gut microbiota metabolism-related arachidonic acid metabolic pathway contributed to miR-30a-5p-regulated hepatic steatosis and lipid disorder. Reintroducing miR-30a-5p reversed hepatic steatosis and arachidonic acid metabolism disorder caused by HFD and miR-30a-5p deletion.
Collapse
Affiliation(s)
- Ruiying Wang
- Xiamen Cardiovascular Hospital of Xiamen UniversitySchool of Medicine, Xiamen UniversityXiamenFujianP. R. China
- Xiamen Key Laboratory of Cardiovascular DiseasesXiamen Cardiovascular Hospital of Xiamen UniversityXiamenFujianP. R. China
| | - Xiaocheng Zhang
- Xiamen Cardiovascular Hospital of Xiamen UniversitySchool of Medicine, Xiamen UniversityXiamenFujianP. R. China
| | - Yutian Wang
- Department of CardiologyNanfang HospitalSouthern Medical UniversityGuangzhouGuangdongP. R. China
| | - Yijun Lin
- Xiamen Cardiovascular Hospital of Xiamen UniversitySchool of Medicine, Xiamen UniversityXiamenFujianP. R. China
- Xiamen Key Laboratory of Cardiovascular DiseasesXiamen Cardiovascular Hospital of Xiamen UniversityXiamenFujianP. R. China
| | - Yuling Zhou
- Xiamen Cardiovascular Hospital of Xiamen UniversitySchool of Medicine, Xiamen UniversityXiamenFujianP. R. China
- Xiamen Key Laboratory of Cardiovascular DiseasesXiamen Cardiovascular Hospital of Xiamen UniversityXiamenFujianP. R. China
| | - Yan Wang
- Xiamen Cardiovascular Hospital of Xiamen UniversitySchool of Medicine, Xiamen UniversityXiamenFujianP. R. China
- Xiamen Key Laboratory of Cardiovascular DiseasesXiamen Cardiovascular Hospital of Xiamen UniversityXiamenFujianP. R. China
| | - Gang Li
- Xiamen Cardiovascular Hospital of Xiamen UniversitySchool of Medicine, Xiamen UniversityXiamenFujianP. R. China
- Xiamen Key Laboratory of Cardiovascular DiseasesXiamen Cardiovascular Hospital of Xiamen UniversityXiamenFujianP. R. China
| |
Collapse
|
26
|
Cuervo L, McAlpine PL, Olano C, Fernández J, Lombó F. Low-Molecular-Weight Compounds Produced by the Intestinal Microbiota and Cardiovascular Disease. Int J Mol Sci 2024; 25:10397. [PMID: 39408727 PMCID: PMC11477366 DOI: 10.3390/ijms251910397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 09/20/2024] [Accepted: 09/24/2024] [Indexed: 10/20/2024] Open
Abstract
Cardiovascular disease is the main cause of mortality in industrialized countries, with over 500 million people affected worldwide. In this work, the roles of low-molecular-weight metabolites originating from the gut microbiome, such as short-chain fatty acids, hydrogen sulfide, trimethylamine, phenylacetic acid, secondary bile acids, indoles, different gases, neurotransmitters, vitamins, and complex lipids, are discussed in relation to their CVD-promoting or preventing activities. Molecules of mixed microbial and human hepatic origin, such as trimethylamine N-oxide and phenylacetylglutamine, are also presented. Finally, dietary agents with cardioprotective effects, such as probiotics, prebiotics, mono- and poly-unsaturated fatty acids, carotenoids, and polyphenols, are also discussed. A special emphasis is given to their gut microbiota-modulating properties.
Collapse
Affiliation(s)
- Lorena Cuervo
- Research Group BIOMIC (Biosynthesis of Antitumor Molecules), Departamento de Biología Funcional, Área de Microbiología, Universidad de Oviedo, 33006 Oviedo, Spain; (L.C.); (C.O.)
- IUOPA (Instituto Universitario de Oncología del Principado de Asturias), 33006 Oviedo, Spain
- ISPA (Instituto de Investigación Sanitaria del Principado de Asturias), 33006 Oviedo, Spain
| | - Patrick L. McAlpine
- IUOPA (Instituto Universitario de Oncología del Principado de Asturias), 33006 Oviedo, Spain
- ISPA (Instituto de Investigación Sanitaria del Principado de Asturias), 33006 Oviedo, Spain
- Research Group BIONUC (Biotechnology of Nutraceuticals and Bioactive Compounds), Departamento de Biología Funcional, Área de Microbiología, Universidad de Oviedo, 33006 Oviedo, Spain
| | - Carlos Olano
- Research Group BIOMIC (Biosynthesis of Antitumor Molecules), Departamento de Biología Funcional, Área de Microbiología, Universidad de Oviedo, 33006 Oviedo, Spain; (L.C.); (C.O.)
- IUOPA (Instituto Universitario de Oncología del Principado de Asturias), 33006 Oviedo, Spain
- ISPA (Instituto de Investigación Sanitaria del Principado de Asturias), 33006 Oviedo, Spain
| | - Javier Fernández
- IUOPA (Instituto Universitario de Oncología del Principado de Asturias), 33006 Oviedo, Spain
- ISPA (Instituto de Investigación Sanitaria del Principado de Asturias), 33006 Oviedo, Spain
- Research Group BIONUC (Biotechnology of Nutraceuticals and Bioactive Compounds), Departamento de Biología Funcional, Área de Microbiología, Universidad de Oviedo, 33006 Oviedo, Spain
| | - Felipe Lombó
- IUOPA (Instituto Universitario de Oncología del Principado de Asturias), 33006 Oviedo, Spain
- ISPA (Instituto de Investigación Sanitaria del Principado de Asturias), 33006 Oviedo, Spain
- Research Group BIONUC (Biotechnology of Nutraceuticals and Bioactive Compounds), Departamento de Biología Funcional, Área de Microbiología, Universidad de Oviedo, 33006 Oviedo, Spain
| |
Collapse
|
27
|
Ma K, Sun L, Jia C, Kui H, Xie J, Zang S, Huang S, Que J, Liu C, Huang J. Potential mechanisms underlying podophyllotoxin-induced cardiotoxicity in male rats: toxicological evidence chain (TEC) concept. Front Pharmacol 2024; 15:1378758. [PMID: 39386032 PMCID: PMC11463157 DOI: 10.3389/fphar.2024.1378758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 07/31/2024] [Indexed: 10/12/2024] Open
Abstract
Introduction Podophyllotoxin (PPT) is a high-content and high-activity compound extracted from the traditional Chinese medicinal plant Dysosma versipellis (DV) which exhibits various biological activities. However, its severe toxicity limits its use. In clinical settings, patients with DV poisoning often experience adverse reactions when taking large doses in a short period. The heart is an important toxic target organ, so it is necessary to conduct 24-h acute cardiac toxicity studies on PPT to understand its underlying toxicity mechanism. Methods Based on the concept of the toxicological evidence chain (TEC), we utilized targeted metabolomic and transcriptomic analyses to reveal the mechanism of the acute cardiotoxicity of PPT. The manifestation of toxicity in Sprague-Dawley rats, including changes in weight and behavior, served as Injury Phenotype Evidence (IPE). To determine Adverse Outcomes Evidence (AOE), the hearts of the rats were evaluated through histopathological examination and by measuring myocardial enzyme and cardiac injury markers levels. Additionally, transcriptome analysis, metabolome analysis, myocardial enzymes, and cardiac injury markers were integrated to obtain Toxic Event Evidence (TEE) using correlation analysis. Results The experiment showed significant epistaxis, hypokinesia, and hunched posture in PPT group rats within 24 h after exposure to 120 mg/kg PPT. It is found that PPT induced cardiac injury in rats within 24 h, as evidenced by increased serum myocardial enzyme levels, elevated concentrations of cardiac injury biomarkers, and altered cardiac cell morphology, all indicating some degree of cardiac toxicity. Transcriptome analysis revealed that primary altered metabolic pathway was arachidonic acid metabolism after PPT exposure. Cyp2e1, Aldob were positively correlated with differential metabolites, while DHA showed positive correlation with differential genes Fmo2 and Timd2, as well as with heart injury markers BNP and Mb. Conclusion This study comprehensively evaluated cardiac toxicity of PPT and initially revealed the mechanism of PPT-induced acute cardiotoxicity, which involved oxidative stress, apoptosis, inflammatory response, and energy metabolism disorder.
Collapse
Affiliation(s)
- Kaiyue Ma
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Lu Sun
- College of Chinese Materia Medica and Food Engineering, Shanxi University of Chinese Medicine, Jinzhong, China
| | - Chunxue Jia
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Hongqian Kui
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Jiaqi Xie
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Shidan Zang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Shixin Huang
- Luoyang Key Laboratory of Clinical Multiomics and Translational Medicine, Henan Key Laboratory of Rare Diseases, Endocrinology and Metabolism Center, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Jinfeng Que
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Chuanxin Liu
- Eye Hospital China Academy of Chinese Medical Sciences, Beijing, China
| | - Jianmei Huang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
28
|
Qi L, Li Y, Chen Z, Wei C, Wen X, Hu S, Wu H, Lv Z, Xu Z, Xia L. Microbiome-metabolome analysis insight into the effects of high-salt diet on hemorheological functions in SD rats. Front Nutr 2024; 11:1408778. [PMID: 39381352 PMCID: PMC11460366 DOI: 10.3389/fnut.2024.1408778] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 08/21/2024] [Indexed: 10/10/2024] Open
Abstract
The present study examined the effect of two dietary regimens with elevated salt concentrations (4% and 8% salt) on hemorheological functions of SD rats, and explored the underlying mechanisms mainly through microbiome-metabolome analysis. An 8% HSD substantially altered the hemorheological parameters, and compromised intestinal barrier integrity and reduced the short-chain fatty acid levels. The microbiome-metabolome analysis revealed that 49 genus-specific microorganisms and 156 metabolites showed a consistent trend after exposure to both 4% and 8% HSDs. Pathway analysis identified significant alterations in key metabolites within bile acid and arachidonic acid metabolism pathways. A two-sample Mendelian randomization (MR) analysis verified the link between high dietary salt intake and hemorheology. It also suggested that some key microbes and metabolites (such as Ruminococcaceae_UCG-005, Lachnospiraceae_NK4A136, Ruminiclostridium_6, and Ruminococcaceae_UCG-010, TXB-2, 11,12-diHETrE, glycochenodeoxycholate) may involve in abnormalities in blood rheology caused by high salt intake. Collectively, our findings underscored the adverse effects of high dietary salt on hemorheological functions and provide new insight into the underlying mechanism based on microbiome-metabolome analysis.
Collapse
Affiliation(s)
- Luming Qi
- School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Yao Li
- Jiangxi Province Key Laboratory of Traditional Chinese Medicine Pharmacology, Institute of Traditional Chinese Medicine Health Industry, China Academy of Chinese Medical Sciences, Nanchang, China
| | - Zhixuan Chen
- School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Changhong Wei
- School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Xue Wen
- School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Shuangyan Hu
- School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Hang Wu
- School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Zhuoheng Lv
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Zhangmeng Xu
- Department of Neck, Shoulder, Waist, and Leg Pain, Sichuan Province Orthopedic Hospital, Chengdu, Sichuan, China
| | - Lina Xia
- School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| |
Collapse
|
29
|
Dong Z, Wang Q. The causal relationship between human blood metabolites and risk of peripheral artery disease: a Mendelian randomization study. Front Cardiovasc Med 2024; 11:1435106. [PMID: 39318836 PMCID: PMC11420124 DOI: 10.3389/fcvm.2024.1435106] [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: 05/19/2024] [Accepted: 08/27/2024] [Indexed: 09/26/2024] Open
Abstract
Background Peripheral Artery Disease (PAD) is a common vascular disorder typically caused by atherosclerosis, leading to impaired blood supply to the lower extremities, resulting in pain, necrosis, and even amputation. Despite extensive research into the pathogenesis of PAD, many mysteries remain, particularly regarding its association with human blood metabolites. Methods To explore the causal relationship between 1,400 serum metabolites and PAD, a two-sample Mendelian randomization (MR) analysis was conducted. The Inverse Variance-Weighted (IVW) method was the primary technique used to estimate the causal impact of the metabolites on PAD. To enhance the analysis, several additional methods were employed: MR-Egger regression, weighted median, simple mode, and weighted mode. These methods provided a comprehensive evaluation beyond the primary IVW estimation. To ensure the validity of the MR findings, sensitivity analysis was performed. Furthermore, a bidirectional MR approach was applied to explore the possibility of a reverse causal effect between PAD and potential candidate metabolites. Results After rigorous selection, significant associations were found between 1-(1-enyl-stearoyl)-2-arachidonoyl-GPE (p-18:0/20:4) and X-17653 levels with PAD. 1-(1-enyl-stearoyl)-2-arachidonoyl-GPE (p-18:0/20:4) was positively associated with increased PAD risk (IVW OR = 1.13, 95% CI, 1.06-1.21; P < 0.001). X-17653 levels were associated with decreased PAD risk (IVW OR = 0.88, 95% CI, 0.83-0.94; P < 0.001). In the reverse direction, PAD was positively associated with increased 1-(1-enyl-stearoyl)-2-arachidonoyl-GPE (p-18:0/20:4) levels (IVW OR = 1.16, 95% CI, 1.01-1.34; P = 0.036). PAD was not associated with X-17653. Conclusion Among 1,400 blood metabolites, 1-(1-enyl-stearoyl)-2-arachidonoyl-GPE (p-18:0/20:4) and X-17653 are significantly associated with PAD risk. Importantly, in the reverse direction, PAD was found to be positively associated with increased levels of 1-(1-enyl-stearoyl)-2-arachidonoyl-GPE (p-18:0/20:4). This highlights the bidirectional nature of the association and suggests a potential feedback mechanism between PAD and this specific lipid species. 1-(1-enyl-stearoyl)-2-arachidonoyl-GPE (p-18:0/20:4) may serve as potential biomarkers for PAD, aiding early diagnosis and providing novel avenues for personalized treatment and management. However, further validation and research are warranted despite the promising results.
Collapse
Affiliation(s)
| | - Qingyun Wang
- Department of Cardiothoracic Surgery, Beijing Shunyi Hospital, Beijing, China
| |
Collapse
|
30
|
Vass Z, Shenker-Horváth K, Bányai B, Vető KN, Török V, Gém JB, Nádasy GL, Kovács KB, Horváth EM, Jakus Z, Hunyady L, Szekeres M, Dörnyei G. Investigating the Role of Cannabinoid Type 1 Receptors in Vascular Function and Remodeling in a Hypercholesterolemic Mouse Model with Low-Density Lipoprotein-Cannabinoid Type 1 Receptor Double Knockout Animals. Int J Mol Sci 2024; 25:9537. [PMID: 39273484 PMCID: PMC11395437 DOI: 10.3390/ijms25179537] [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: 07/16/2024] [Revised: 08/24/2024] [Accepted: 08/26/2024] [Indexed: 09/15/2024] Open
Abstract
Hypercholesterolemia forms the background of several cardiovascular pathologies. LDL receptor-knockout (LDLR-KO) mice kept on a high-fat diet (HFD) develop high cholesterol levels and atherosclerosis (AS). Cannabinoid type 1 receptors (CB1Rs) induce vasodilation, although their role in cardiovascular pathologies is still controversial. We aimed to reveal the effects of CB1Rs on vascular function and remodeling in hypercholesterolemic AS-prone LDLR-KO mice. Experiments were performed on a newly established LDLR and CB1R double-knockout (KO) mouse model, in which KO and wild-type (WT) mice were kept on an HFD or a control diet (CD) for 5 months. The vascular functions of abdominal aorta rings were tested with wire myography. The vasorelaxation effects of acetylcholine (Ach, 1 nM-1 µM) were obtained after phenylephrine precontraction, which was repeated with inhibitors of nitric oxide synthase (NOS) and cyclooxygenase (COX), Nω-nitro-L-arginine (LNA), and indomethacin (INDO), respectively. Blood pressure was measured with the tail-cuff method. Immunostaining of endothelial NOS (eNOS) was carried out. An HFD significantly elevated the cholesterol levels in the LDLR-KO mice more than in the corresponding WT mice (mean values: 1039 ± 162 mg/dL vs. 91 ± 18 mg/dL), and they were not influenced by the presence of the CB1R gene. However, with the defect of the CB1R gene, damage to the Ach relaxation ability was moderated. The blood pressure was higher in the LDLR-KO mice compared to their WT counterparts (systolic/diastolic values: 110/84 ± 5.8/6.8 vs. 102/80 ± 3.3/2.5 mmHg), which was significantly elevated with an HFD (118/96 ± 1.9/2 vs. 100/77 ± 3.4/3.1 mmHg, p < 0.05) but attenuated in the CB1R-KO HFD mice. The expression of eNOS was depressed in the HFD WT mice compared to those on the CD, but it was augmented if CB1R was knocked out. This newly established double-knockout mouse model provides a tool for studying the involvement of CB1Rs in the development of hypercholesterolemia and atherosclerosis. Our results indicate that knocking out the CB1R gene significantly attenuates vascular damage in hypercholesterolemic mice.
Collapse
MESH Headings
- Animals
- Male
- Mice
- Acetylcholine/pharmacology
- Atherosclerosis/metabolism
- Atherosclerosis/genetics
- Atherosclerosis/pathology
- Atherosclerosis/etiology
- Diet, High-Fat/adverse effects
- Disease Models, Animal
- Hypercholesterolemia/metabolism
- Hypercholesterolemia/genetics
- Hypercholesterolemia/pathology
- Mice, Inbred C57BL
- Mice, Knockout
- Nitric Oxide Synthase Type III/metabolism
- Receptor, Cannabinoid, CB1/metabolism
- Receptor, Cannabinoid, CB1/genetics
- Receptors, LDL/genetics
- Receptors, LDL/metabolism
- Receptors, LDL/deficiency
- Vascular Remodeling/drug effects
- Vasodilation/drug effects
Collapse
Affiliation(s)
- Zsolt Vass
- Department of Morphology and Physiology, Faculty of Health Sciences, Semmelweis University, 17 Vas Street, 1088 Budapest, Hungary; (Z.V.); (K.S.-H.); (K.N.V.); (V.T.); (G.D.)
| | - Kinga Shenker-Horváth
- Department of Morphology and Physiology, Faculty of Health Sciences, Semmelweis University, 17 Vas Street, 1088 Budapest, Hungary; (Z.V.); (K.S.-H.); (K.N.V.); (V.T.); (G.D.)
- Center for Sports Nutrition Science, Hungarian University of Sports Science, 42-48 Alkotás Street, 1123 Budapest, Hungary
| | - Bálint Bányai
- Department of Physiology, Faculty of Medicine, Semmelweis University, 37-47 Tűzoltó Street, 1094 Budapest, Hungary; (B.B.); (J.B.G.); (G.L.N.); (K.B.K.); (E.M.H.); (Z.J.); (L.H.)
| | - Kinga Nóra Vető
- Department of Morphology and Physiology, Faculty of Health Sciences, Semmelweis University, 17 Vas Street, 1088 Budapest, Hungary; (Z.V.); (K.S.-H.); (K.N.V.); (V.T.); (G.D.)
| | - Viktória Török
- Department of Morphology and Physiology, Faculty of Health Sciences, Semmelweis University, 17 Vas Street, 1088 Budapest, Hungary; (Z.V.); (K.S.-H.); (K.N.V.); (V.T.); (G.D.)
| | - Janka Borbála Gém
- Department of Physiology, Faculty of Medicine, Semmelweis University, 37-47 Tűzoltó Street, 1094 Budapest, Hungary; (B.B.); (J.B.G.); (G.L.N.); (K.B.K.); (E.M.H.); (Z.J.); (L.H.)
| | - György L. Nádasy
- Department of Physiology, Faculty of Medicine, Semmelweis University, 37-47 Tűzoltó Street, 1094 Budapest, Hungary; (B.B.); (J.B.G.); (G.L.N.); (K.B.K.); (E.M.H.); (Z.J.); (L.H.)
| | - Kinga Bernadett Kovács
- Department of Physiology, Faculty of Medicine, Semmelweis University, 37-47 Tűzoltó Street, 1094 Budapest, Hungary; (B.B.); (J.B.G.); (G.L.N.); (K.B.K.); (E.M.H.); (Z.J.); (L.H.)
| | - Eszter Mária Horváth
- Department of Physiology, Faculty of Medicine, Semmelweis University, 37-47 Tűzoltó Street, 1094 Budapest, Hungary; (B.B.); (J.B.G.); (G.L.N.); (K.B.K.); (E.M.H.); (Z.J.); (L.H.)
| | - Zoltán Jakus
- Department of Physiology, Faculty of Medicine, Semmelweis University, 37-47 Tűzoltó Street, 1094 Budapest, Hungary; (B.B.); (J.B.G.); (G.L.N.); (K.B.K.); (E.M.H.); (Z.J.); (L.H.)
| | - László Hunyady
- Department of Physiology, Faculty of Medicine, Semmelweis University, 37-47 Tűzoltó Street, 1094 Budapest, Hungary; (B.B.); (J.B.G.); (G.L.N.); (K.B.K.); (E.M.H.); (Z.J.); (L.H.)
- Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, 2 Magyar Tudósok Körútja, 1117 Budapest, Hungary
| | - Mária Szekeres
- Department of Morphology and Physiology, Faculty of Health Sciences, Semmelweis University, 17 Vas Street, 1088 Budapest, Hungary; (Z.V.); (K.S.-H.); (K.N.V.); (V.T.); (G.D.)
- Department of Physiology, Faculty of Medicine, Semmelweis University, 37-47 Tűzoltó Street, 1094 Budapest, Hungary; (B.B.); (J.B.G.); (G.L.N.); (K.B.K.); (E.M.H.); (Z.J.); (L.H.)
| | - Gabriella Dörnyei
- Department of Morphology and Physiology, Faculty of Health Sciences, Semmelweis University, 17 Vas Street, 1088 Budapest, Hungary; (Z.V.); (K.S.-H.); (K.N.V.); (V.T.); (G.D.)
| |
Collapse
|
31
|
Sun S, Hara A, Johnstone L, Hallmark B, Watkins JC, Thomson CA, Schembre SM, Sergeant S, Umans JG, Yao G, Zhang HH, Chilton FH. Optimal Pair Matching Combined with Machine Learning Predicts a Significant Reduction in Myocardial Infarction Risk in African Americans Following Omega-3 Fatty Acid Supplementation. Nutrients 2024; 16:2933. [PMID: 39275249 PMCID: PMC11397525 DOI: 10.3390/nu16172933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 08/16/2024] [Accepted: 08/24/2024] [Indexed: 09/16/2024] Open
Abstract
Conflicting clinical trial results on omega-3 highly unsaturated fatty acids (n-3 HUFA) have prompted uncertainty about their cardioprotective effects. While the VITAL trial found no overall cardiovascular benefit from n-3 HUFA supplementation, its substantial African American (AfAm) enrollment provided a unique opportunity to explore racial differences in response to n-3 HUFA supplementation. The current observational study aimed to simulate randomized clinical trial (RCT) conditions by matching 3766 AfAm and 15,553 non-Hispanic White (NHW) individuals from the VITAL trial utilizing propensity score matching to address the limitations related to differences in confounding variables between the two groups. Within matched groups (3766 AfAm and 3766 NHW), n-3 HUFA supplementation's impact on myocardial infarction (MI), stroke, and cardiovascular disease (CVD) mortality was assessed. A weighted decision tree analysis revealed belonging to the n-3 supplementation group as the most significant predictor of MI among AfAm but not NHW. Further logistic regression using the LASSO method and bootstrap estimation of standard errors indicated n-3 supplementation significantly lowered MI risk in AfAm (OR 0.17, 95% CI [0.048, 0.60]), with no such effect in NHW. This study underscores the critical need for future RCT to explore racial disparities in MI risk associated with n-3 HUFA supplementation and highlights potential causal differences between supplementation health outcomes in AfAm versus NHW populations.
Collapse
Affiliation(s)
- Shudong Sun
- Department of Mathematics, University of Arizona, Tucson, AZ 85721, USA
- Statistics Interdisciplinary Program, University of Arizona, Tucson, AZ 85721, USA
| | - Aki Hara
- School of Nutritional Sciences and Wellness, College of Agriculture and Life Sciences, University of Arizona, Tucson, AZ 85719, USA
| | - Laurel Johnstone
- School of Nutritional Sciences and Wellness, College of Agriculture and Life Sciences, University of Arizona, Tucson, AZ 85719, USA
| | - Brian Hallmark
- BIO5 Institute, University of Arizona, Tucson, AZ 85719, USA
- Center for Precision Nutrition and Wellness, University of Arizona, Tucson, AZ 85719, USA
| | - Joseph C Watkins
- Department of Mathematics, University of Arizona, Tucson, AZ 85721, USA
- Statistics Interdisciplinary Program, University of Arizona, Tucson, AZ 85721, USA
| | - Cynthia A Thomson
- Department of Health Promotion Sciences, Mel & Enid Zuckerman College of Public Health, University of Arizona, Tucson, AZ 85724, USA
| | - Susan M Schembre
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20007, USA
| | - Susan Sergeant
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Jason G Umans
- Georgetown-Howard Universities Center for Clinical and Translational Science, Washington, DC 20057, USA
| | - Guang Yao
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, AZ 85721, USA
| | - Hao Helen Zhang
- Department of Mathematics, University of Arizona, Tucson, AZ 85721, USA
- Statistics Interdisciplinary Program, University of Arizona, Tucson, AZ 85721, USA
| | - Floyd H Chilton
- School of Nutritional Sciences and Wellness, College of Agriculture and Life Sciences, University of Arizona, Tucson, AZ 85719, USA
- BIO5 Institute, University of Arizona, Tucson, AZ 85719, USA
- Center for Precision Nutrition and Wellness, University of Arizona, Tucson, AZ 85719, USA
- Escuela de Medicina y Ciencias de la Salud, Tecnologico de Monterrey, Monterrey 64710, Mexico
| |
Collapse
|
32
|
Wang L, Sun Y, Du L, Wang Q, Zhan M, Li S, Xiao X. Daily koumiss has positive regulatory effects on blood lipids and immune system: A metabolomics study. Heliyon 2024; 10:e36429. [PMID: 39253138 PMCID: PMC11382052 DOI: 10.1016/j.heliyon.2024.e36429] [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: 11/04/2023] [Revised: 08/05/2024] [Accepted: 08/15/2024] [Indexed: 09/11/2024] Open
Abstract
Koumiss, a traditional Mongolian beverage, is believed to possess high nutritional value and potential medical benefits. However, there is a lack of comprehensive research on its potential impact on the human body. Metabolomics, as a sensitive approach in systems biology, offers a new avenue for studying the overall effects of koumiss. In this work, metabolomics was utilized to identify potential biomarkers and pathways associated with koumiss using UPLC-MS detection, pattern recognition analysis, pathway enrichment, network pharmacology. The findings indicated that koumiss exerts a beneficial regulatory influence on lipid metabolism, neurotransmitters, hormones, phospholipids and arachidonic acid metabolism, besides up regulating the content of nutrients. It could reduce the risks of dyslipidemia and inflammatory responses. This study confirmed the benign regulatory effect of koumiss on normal organism from the perspective of endogenous metabolites, and provided objective support for the promotion and application of this ethnic food.
Collapse
Affiliation(s)
- Leqi Wang
- Institute of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yuanfang Sun
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Lijing Du
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Qian Wang
- Institute of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Min Zhan
- Institute of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shasha Li
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xue Xiao
- Institute of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| |
Collapse
|
33
|
Zhang Y, Zhang Y, Yin R, Fang X, Miao R, Guan H, Yao Y, Tian J. Multi-omics characterization of type 2 diabetes mellitus-induced gastroenteropathy in the db/db mouse model. Front Cell Dev Biol 2024; 12:1417255. [PMID: 39211388 PMCID: PMC11357919 DOI: 10.3389/fcell.2024.1417255] [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: 04/14/2024] [Accepted: 08/05/2024] [Indexed: 09/04/2024] Open
Abstract
Objective Gastrointestinal dysfunction are often associated with type 2 diabetes mellitus (T2DM), a complicated metabolic illness. Contributing factors have been proposed, including genetic predisposition, gene environmental, and lifestyle interactions, but the pathophysiology remains unknown. Methods We aim to explore the possible causes behind gastrointestinal dysfunction caused by type 2 diabetes in this study. A comprehensive analysis of the gastric sinus metabolome, transcriptome, and proteome in db/db mice with gastrointestinal dysfunction was conducted. Results The model group of mice had considerably lower small intestine propulsion and gastric emptying rates, higher blood glucose levels, and were significantly obese compared to the control group. We identified 297 genes, 350 proteins, and 1,001 metabolites exhibiting significant differences between db/db and control mice (p < 0.05). Moreover, multi-omics analysis revealed that the genes, proteins, and metabolites in the T2DM-induced gastroenteropathy mice group were involved in arachidonic acid metabolism, glycerophospholipid metabolism and vitamin digestion and absorption. Specifically, Cbr3, Etnppl, and Apob were the major mRNAs associated with T2DM-induced gastrointestinal dysfunction, while Cyp2b10, Cyp2b19, Pgs1, Gpat3, Apoa4, and Tcn2 were the major proteins associated with T2DM-induced gastrointestinal injury, and 16(R)-HET, 5-HETE, LysoPC (22:0), and Pantothenic acid were the major metabolites associated with T2DM-induced gastrointestinal disorders. Conclusion The mechanism of action of diabetic gastroenteropathy may be related to vitamin digestion and absorption, glycerophospholipid metabolism, and arachidonic acid metabolism.
Collapse
Affiliation(s)
- Yuxin Zhang
- Institute of Metabolic Diseases, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yanjiao Zhang
- Institute of Metabolic Diseases, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ruiyang Yin
- Institute of Metabolic Diseases, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xinyi Fang
- Institute of Metabolic Diseases, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate College, Beijing University of Chinese Medicine, Beijing, China
| | - Runyu Miao
- Institute of Metabolic Diseases, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate College, Beijing University of Chinese Medicine, Beijing, China
| | - Huifang Guan
- Graduate College, Changchun University of Chinese Medicine, Changchun, China
| | - Yiqi Yao
- Institute of Metabolic Diseases, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jiaxing Tian
- Institute of Metabolic Diseases, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
34
|
Helal SA, El-Sherbeni AA, El-Kadi AOS. 11-Hydroxyeicosatetraenoics induces cellular hypertrophy in an enantioselective manner. Front Pharmacol 2024; 15:1438567. [PMID: 39188949 PMCID: PMC11345585 DOI: 10.3389/fphar.2024.1438567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Accepted: 07/08/2024] [Indexed: 08/28/2024] Open
Abstract
Background R/S enantiomers of 11-hydroxyeicosatertraenoic acid (11-HETE) are formed from arachidonic acid by enzymatic and non-enzymatic pathways. 11-HETE is predominately formed by the cytochrome P450 1B1 (CYP1B1). The role of CYP1B1 in the development of cardiovascular diseases is well established. Objectives This study aimed to assess the cellular hypertrophic effect of 11-HETE enantiomers in human RL-14 cardiomyocyte cell line and to examine their association with CYP1B1 levels. Methods Human fetal ventricular cardiomyocyte, RL-14 cells, were treated with 20 µM (R) or (S) 11-HETE for 24 h. Thereafter, cellular hypertrophic markers and cell size were then determined using real-time polymerase chain reaction (RT-PCR) and phase-contrast imaging, respectively. The mRNA and protein levels of selected CYPs were determined using RT-PCR and Western blot, respectively. In addition, we examined the effect of (R) and (S) 11-HETE on CYP1B1 catalytic activity using human recombinant CYP1B1 and human liver microsomes. Results Both (R) and (S) 11-HETE induced cellular hypertrophic markers and cell surface area in RL-14 cells. Both enantiomers significantly upregulated CYP1B1, CYP1A1, CYP4F2, and CYP4A11 at the mRNA and protein levels, however, the effect of the S-enantiomer was more pronounced. Furthermore, 11(S)-HETE increased the mRNA and protein levels of CYP2J and CYP4F2, whereas 11(R)-HETE increased only CYP4F2. Only 11(S)-HETE significantly increased the catalytic activity of CYP1B1 in recombinant human CYP1B1, suggesting allosteric activation in an enantioselective manner. Conclusion Our study provides the first evidence that 11-HETE can induce cellular hypertrophy in RL-14 cells via the increase in CYP1B1 mRNA, protein, and activity levels.
Collapse
Affiliation(s)
- Sara A. Helal
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
- Department of Biochemistry, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Ahmed A. El-Sherbeni
- Department of Clinical Pharmacy, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Ayman O. S. El-Kadi
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
35
|
Xu L, Yang Q, Zhou J. Mechanisms of Abnormal Lipid Metabolism in the Pathogenesis of Disease. Int J Mol Sci 2024; 25:8465. [PMID: 39126035 PMCID: PMC11312913 DOI: 10.3390/ijms25158465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 07/26/2024] [Accepted: 07/31/2024] [Indexed: 08/12/2024] Open
Abstract
Lipid metabolism is a critical component in preserving homeostasis and health, and lipids are significant chemicals involved in energy metabolism in living things. With the growing interest in lipid metabolism in recent years, an increasing number of studies have demonstrated the close relationship between abnormalities in lipid metabolism and the development of numerous human diseases, including cancer, cardiovascular, neurological, and endocrine system diseases. Thus, understanding how aberrant lipid metabolism contributes to the development of related diseases and how it works offers a theoretical foundation for treating and preventing related human diseases as well as new avenues for the targeted treatment of related diseases. Therefore, we discuss the processes of aberrant lipid metabolism in various human diseases in this review, including diseases of the cardiovascular system, neurodegenerative diseases, endocrine system diseases (such as obesity and type 2 diabetes mellitus), and other diseases including cancer.
Collapse
Affiliation(s)
| | | | - Jinghua Zhou
- School of Basic Medicine Sciences, Hangzhou Normal University, Hangzhou 311121, China
| |
Collapse
|
36
|
Gerges SH, El-Kadi AOS. Changes in cardiovascular arachidonic acid metabolism in experimental models of menopause and implications on postmenopausal cardiac hypertrophy. Prostaglandins Other Lipid Mediat 2024; 173:106851. [PMID: 38740361 DOI: 10.1016/j.prostaglandins.2024.106851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 04/17/2024] [Accepted: 05/07/2024] [Indexed: 05/16/2024]
Abstract
Menopause is a normal stage in the human female aging process characterized by the cessation of menstruation and the ovarian production of estrogen and progesterone hormones. Menopause is associated with an increased risk of several different diseases. Cardiovascular diseases are generally less common in females than in age-matched males. However, this female advantage is lost after menopause. Cardiac hypertrophy is a disease characterized by increased cardiac size that develops as a response to chronic overload or stress. Similar to other cardiovascular diseases, the risk of cardiac hypertrophy significantly increases after menopause. However, the exact underlying mechanisms are not yet fully elucidated. Several studies have shown that surgical or chemical induction of menopause in experimental animals is associated with cardiac hypertrophy, or aggravates cardiac hypertrophy induced by other stressors. Arachidonic acid (AA) released from the myocardial phospholipids is metabolized by cardiac cytochrome P450 (CYP), cyclooxygenase (COX), and lipoxygenase (LOX) enzymes to produce several eicosanoids. AA-metabolizing enzymes and their respective metabolites play an important role in the pathogenesis of cardiac hypertrophy. Menopause is associated with changes in the cardiovascular levels of CYP, COX, and LOX enzymes and the levels of their metabolites. It is possible that these changes might play a role in the increased risk of cardiac hypertrophy after menopause.
Collapse
Affiliation(s)
- Samar H Gerges
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Ayman O S El-Kadi
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
37
|
Alammari AH, Isse FA, O'Croinin C, Davies NM, El-Kadi AOS. Modulation of Angiotensin II-Induced Cellular Hypertrophy by Cannflavin-C: Unveiling the Impact on Cytochrome P450 1B1 and Arachidonic Acid Metabolites. Drug Metab Dispos 2024; 52:875-885. [PMID: 38839111 DOI: 10.1124/dmd.124.001705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 05/23/2024] [Accepted: 05/29/2024] [Indexed: 06/07/2024] Open
Abstract
This research aimed to clarify the impacts of cannflavin-C on angiotensin II (Ang II)-induced cardiac hypertrophy and their potential role in modulating cytochrome P450 1B1 (CYP1B1) and arachidonic acid (AA) metabolites. Currently there is no evidence to suggest that cannflavin-C, a prenylated flavonoid, has any significant effects on the heart or cardiac hypertrophy. The metabolism of arachidonic acid (AA) into midchain hydroxyeicosatetraenoic acids (HETEs), facilitated by CYP1B1 enzyme, plays a role in the development of cardiac hypertrophy, which is marked by enlarged cardiac cells. Adult human ventricular cardiomyocyte (AC16) cell line was cultured and exposed to cannflavin-C in the presence and absence of Ang II. The assessment of mRNA expression pertaining to cardiac hypertrophic markers and cytochromes P450 (P450s) was conducted via real-time polymerase chain reaction (PCR), whereas the quantification of P450 protein levels was carried out through western blot analysis. Ang II induced hypertrophic markers myosin heavy chain (β/α-MHC), atrial natriuretic peptide (ANP), and brain natriuretic peptide (BNP) and increased cell surface area, whereas cannflavin-C mitigated these effects. Gene and protein expression analysis revealed that cannflavin-C downregulated CYP1B1 gene expression, protein level, and enzyme activity assessed by 7-methoxyresorufin O-deethylase (MROD). Arachidonic acid metabolites analysis, using liquid chromatography-tandem mass spectrometry (LC-MS/MS), demonstrated that Ang II increased midchain (R/S)-HETE concentrations, which were attenuated by cannflavin-C. This study provides novel insights into the potential of cannflavin-C in modulating arachidonic acid metabolites and attenuating Ang II-induced cardiac hypertrophy, highlighting the importance of this compound as potential therapeutic agents for cardiac hypertrophy. SIGNIFICANCE STATEMENT: This study demonstrates that cannflavin-C offers protection against cellular hypertrophy induced by angiotensin II. The significance of this research lies in its novel discovery, which elucidates a mechanistic pathway involving the inhibition of CYP1B1 by cannflavin-C. This discovery opens up new avenues for leveraging this compound in the treatment of heart failure.
Collapse
Affiliation(s)
- Ahmad H Alammari
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Fadumo Ahmed Isse
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Conor O'Croinin
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Neal M Davies
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Ayman O S El-Kadi
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
38
|
Mu G, Cao X, Shao L, Shen H, Guo X, Gao Y, Su C, Fan H, Yu Y, Shen Z. Progress and perspectives of metabolic biomarkers in human aortic dissection. Metabolomics 2024; 20:76. [PMID: 39002042 DOI: 10.1007/s11306-024-02140-z] [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: 02/02/2024] [Accepted: 06/06/2024] [Indexed: 07/15/2024]
Abstract
BACKGROUND Aortic dissection (AD) significantly threated human cardiovascular health, extensive clinical-scientific research programs have been executed to uncover the pathogenesis and prevention. Unfortunately, no specific biomarker was identified for the causality or development of human AD. AIM OF REVIEW Metabolomics, a high-throughput technique capable of quantitatively detecting metabolites, holds considerable promise in discovering specific biomarkers and unraveling the underlying pathways involved. Aiming to provide a metabolite prediction in human AD, we collected the metabolomics data from 2003 to 2023, and diligently scrutinized with the online system MetaboAnalyst 6.0. KEY SCIENTIFIC CONCEPTS OF REVIEW Based on the data obtained, we have concluded the metabolic dynamics were highly correlated with human AD. Such metabolites (choline, serine and uridine) were frequently involved in the AD. Besides, the pathways, including amino acids metabolism and lipids metabolism, were also dysregulated in the disease. Due to the current limitation of metabolism analysis, the integrative omics data including genomics, transcriptomics, and proteomics were required for developing the specific biomarker for AD.
Collapse
Affiliation(s)
- Gaohang Mu
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, Suzhou, 215123, Jiangsu, China
- Suzhou Medical College, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Xiangyu Cao
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, Suzhou, 215123, Jiangsu, China
- Suzhou Medical College, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Lianbo Shao
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, Suzhou, 215123, Jiangsu, China
- Suzhou Medical College, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Han Shen
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, Suzhou, 215123, Jiangsu, China
- Suzhou Medical College, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Xingyou Guo
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, Suzhou, 215123, Jiangsu, China
- Suzhou Medical College, Soochow University, Suzhou, 215123, Jiangsu, China
- Department of Vascular Surgery, Suqian First Hospital, Suqian, 223800, Jiangsu, China
| | - Yamei Gao
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, Suzhou, 215123, Jiangsu, China
- Suzhou Medical College, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Chengkai Su
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, Suzhou, 215123, Jiangsu, China
- Suzhou Medical College, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Hongyou Fan
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, Suzhou, 215123, Jiangsu, China
- Suzhou Medical College, Soochow University, Suzhou, 215123, Jiangsu, China
| | - You Yu
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, Suzhou, 215123, Jiangsu, China.
- Suzhou Medical College, Soochow University, Suzhou, 215123, Jiangsu, China.
| | - Zhenya Shen
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, Suzhou, 215123, Jiangsu, China.
- Suzhou Medical College, Soochow University, Suzhou, 215123, Jiangsu, China.
| |
Collapse
|
39
|
Lu G, Gao D, Jiang W, Yu X, Tong J, Liu X, Qiao T, Wang R, Zhang M, Wang S, Yang J, Li D, Lv Z. Disrupted gut microecology after high-dose 131I therapy and radioprotective effects of arachidonic acid supplementation. Eur J Nucl Med Mol Imaging 2024; 51:2395-2408. [PMID: 38561516 PMCID: PMC11178657 DOI: 10.1007/s00259-024-06688-9] [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: 01/23/2024] [Accepted: 03/10/2024] [Indexed: 04/04/2024]
Abstract
BACKGROUND Despite the potential radiotoxicity in differentiated thyroid cancer (DTC) patients with high-dose 131I therapy, the alterations and regulatory mechanisms dependent on intestinal microecology remain poorly understood. We aimed to identify the characteristics of the gut microbiota and metabolites in DTC patients suffering from high-dose 131I therapy and explore the radioprotective mechanisms underlying arachidonic acid (ARA) treatment. METHODS A total of 102 patients with DTC were recruited, with fecal samples collected before and after 131I therapy for microbiome and untargeted and targeted metabolomic analyses. Mice were exposed to total body irradiation with ARA replenishment and antibiotic pretreatment and were subjected to metagenomic, metabolomic, and proteomic analyses. RESULTS 131I therapy significantly changed the structure of gut microbiota and metabolite composition in patients with DTC. Lachnospiraceae were the most dominant bacteria after 131I treatment, and metabolites with decreased levels and pathways related to ARA and linoleic acid were observed. In an irradiation mouse model, ARA supplementation not only improved quality of life and recovered hematopoietic and gastrointestinal systems but also ameliorated oxidative stress and inflammation and preserved enteric microecology composition. Additionally, antibiotic intervention eliminated the radioprotective effects of ARA. Proteomic analysis and ursolic acid pretreatment showed that ARA therapy greatly influenced intestinal lipid metabolism in mice subjected to irradiation by upregulating the expression of hydroxy-3-methylglutaryl-coenzyme A synthase 1. CONCLUSION These findings highlight that ARA, as a key metabolite, substantially contributes to radioprotection. Our study provides novel insights into the pivotal role that the microbiota-metabolite axis plays in radionuclide protection and offers effective biological targets for treating radiation-induced adverse effects.
Collapse
Affiliation(s)
- Ganghua Lu
- Clinical Nuclear Medicine Center, Imaging Clinical Medical Center, Institute of Nuclear Medicine, Department of Nuclear Medicine, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
| | - Dingwei Gao
- Clinical Nuclear Medicine Center, Imaging Clinical Medical Center, Institute of Nuclear Medicine, Department of Nuclear Medicine, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
| | - Wen Jiang
- Clinical Nuclear Medicine Center, Imaging Clinical Medical Center, Institute of Nuclear Medicine, Department of Nuclear Medicine, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
| | - Xiaqing Yu
- Clinical Nuclear Medicine Center, Imaging Clinical Medical Center, Institute of Nuclear Medicine, Department of Nuclear Medicine, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
| | - Junyu Tong
- Clinical Nuclear Medicine Center, Imaging Clinical Medical Center, Institute of Nuclear Medicine, Department of Nuclear Medicine, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
| | - Xiaoyan Liu
- Clinical Nuclear Medicine Center, Imaging Clinical Medical Center, Institute of Nuclear Medicine, Department of Nuclear Medicine, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
| | - Tingting Qiao
- Clinical Nuclear Medicine Center, Imaging Clinical Medical Center, Institute of Nuclear Medicine, Department of Nuclear Medicine, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
| | - Ru Wang
- Clinical Nuclear Medicine Center, Imaging Clinical Medical Center, Institute of Nuclear Medicine, Department of Nuclear Medicine, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
| | - Mengyu Zhang
- Clinical Nuclear Medicine Center, Imaging Clinical Medical Center, Institute of Nuclear Medicine, Department of Nuclear Medicine, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
| | - Shaoping Wang
- Clinical Nuclear Medicine Center, Imaging Clinical Medical Center, Institute of Nuclear Medicine, Department of Nuclear Medicine, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
| | - Jianshe Yang
- Clinical Nuclear Medicine Center, Imaging Clinical Medical Center, Institute of Nuclear Medicine, Department of Nuclear Medicine, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China.
| | - Dan Li
- Department of Nuclear Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510289, China.
| | - Zhongwei Lv
- Clinical Nuclear Medicine Center, Imaging Clinical Medical Center, Institute of Nuclear Medicine, Department of Nuclear Medicine, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China.
| |
Collapse
|
40
|
Zhang M, Guo M, Gao Y, Wu C, Pan X, Huang Z. Mechanisms and therapeutic targets of ferroptosis: Implications for nanomedicine design. J Pharm Anal 2024; 14:100960. [PMID: 39135963 PMCID: PMC11318476 DOI: 10.1016/j.jpha.2024.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 02/27/2024] [Accepted: 03/04/2024] [Indexed: 08/15/2024] Open
Abstract
Ferroptosis is a nonapoptotic form of cell death and differs considerably from the well-known forms of cell death in terms of cell morphology, genetics, and biochemistry. The three primary pathways for cell ferroptosis are system Xc-/glutathione peroxidase 4 (GPX4), lipid metabolism, and ferric metabolism. Since the discovery of ferroptosis, mounting evidence has revealed its critical regulatory role in several diseases, especially as a novel potential target for cancer therapy, thereby attracting increasing attention in the fields of tumor biology and anti-tumor therapy. Accordingly, broad prospects exist for identifying ferroptosis as a potential therapeutic target. In this review, we aimed to systematically summarize the activation and defense mechanisms of ferroptosis, highlight the therapeutic targets, and discuss the design of nanomedicines for ferroptosis regulation. In addition, we opted to present the advantages and disadvantages of current ferroptosis research and provide an optimistic vision of future directions in related fields. Overall, we aim to provide new ideas for further ferroptosis research and inspire new strategies for disease diagnosis and treatment.
Collapse
Affiliation(s)
- Meihong Zhang
- College of Pharmacy, University of Jinan, Guangzhou, 510632, China
| | - Mengqin Guo
- College of Pharmacy, University of Jinan, Guangzhou, 510632, China
| | - Yue Gao
- College of Pharmacy, University of Jinan, Guangzhou, 510632, China
| | - Chuanbin Wu
- College of Pharmacy, University of Jinan, Guangzhou, 510632, China
| | - Xin Pan
- College of Pharmacy, University of Sun Yat-sen, Guangzhou, 510275, China
| | - Zhengwei Huang
- College of Pharmacy, University of Jinan, Guangzhou, 510632, China
| |
Collapse
|
41
|
Hu Y, Li W, Cheng X, Yang H, She ZG, Cai J, Li H, Zhang XJ. Emerging Roles and Therapeutic Applications of Arachidonic Acid Pathways in Cardiometabolic Diseases. Circ Res 2024; 135:222-260. [PMID: 38900855 DOI: 10.1161/circresaha.124.324383] [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] [Indexed: 06/22/2024]
Abstract
Cardiometabolic disease has become a major health burden worldwide, with sharply increasing prevalence but highly limited therapeutic interventions. Emerging evidence has revealed that arachidonic acid derivatives and pathway factors link metabolic disorders to cardiovascular risks and intimately participate in the progression and severity of cardiometabolic diseases. In this review, we systemically summarized and updated the biological functions of arachidonic acid pathways in cardiometabolic diseases, mainly focusing on heart failure, hypertension, atherosclerosis, nonalcoholic fatty liver disease, obesity, and diabetes. We further discussed the cellular and molecular mechanisms of arachidonic acid pathway-mediated regulation of cardiometabolic diseases and highlighted the emerging clinical advances to improve these pathological conditions by targeting arachidonic acid metabolites and pathway factors.
Collapse
Affiliation(s)
- Yufeng Hu
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China (Y.H., X.C., H.Y., Z.-G.S., J.C., H.L., X.-J.Z.)
- Key Laboratory of Cardiovascular Disease Prevention and Control, Ministry of Education, First Affiliated Hospital of Gannan Medical University, Ganzhou, China (Y.H., X.C., H.Y.)
| | - Wei Li
- Department of Cardiology, Renmin Hospital of Wuhan University, China (W.L., Z.-G.S., H.L.)
| | - Xu Cheng
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China (Y.H., X.C., H.Y., Z.-G.S., J.C., H.L., X.-J.Z.)
- Key Laboratory of Cardiovascular Disease Prevention and Control, Ministry of Education, First Affiliated Hospital of Gannan Medical University, Ganzhou, China (Y.H., X.C., H.Y.)
| | - Hailong Yang
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China (Y.H., X.C., H.Y., Z.-G.S., J.C., H.L., X.-J.Z.)
- Key Laboratory of Cardiovascular Disease Prevention and Control, Ministry of Education, First Affiliated Hospital of Gannan Medical University, Ganzhou, China (Y.H., X.C., H.Y.)
| | - Zhi-Gang She
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China (Y.H., X.C., H.Y., Z.-G.S., J.C., H.L., X.-J.Z.)
- Department of Cardiology, Renmin Hospital of Wuhan University, China (W.L., Z.-G.S., H.L.)
| | - Jingjing Cai
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China (Y.H., X.C., H.Y., Z.-G.S., J.C., H.L., X.-J.Z.)
- Department of Cardiology, The Third Xiangya Hospital, Central South University, Changsha, China (J.C.)
| | - Hongliang Li
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China (Y.H., X.C., H.Y., Z.-G.S., J.C., H.L., X.-J.Z.)
- Department of Cardiology, Renmin Hospital of Wuhan University, China (W.L., Z.-G.S., H.L.)
- Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, China (H.L.)
| | - Xiao-Jing Zhang
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China (Y.H., X.C., H.Y., Z.-G.S., J.C., H.L., X.-J.Z.)
- School of Basic Medical Sciences, Wuhan University, China (X.-J.Z.)
| |
Collapse
|
42
|
Yan X, Zhang A, Guan Y, Jiao J, Ghanim M, Zhang Y, He X, Shi R. Comparative Metabolome and Transcriptome Analyses Reveal Differential Enrichment of Metabolites with Age in Panax notoginseng Roots. PLANTS (BASEL, SWITZERLAND) 2024; 13:1441. [PMID: 38891250 PMCID: PMC11175106 DOI: 10.3390/plants13111441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 05/05/2024] [Accepted: 05/13/2024] [Indexed: 06/21/2024]
Abstract
Panax notoginseng is a perennial plant well known for its versatile medicinal properties, including hepatoprotective, antioxidant, anti-inflammatory, anti-tumor, estrogen-like, and antidepressant characteristics. It has been reported that plant age affects the quality of P. notoginseng. This study aimed to explore the differential metabolome and transcriptome of 2-year (PN2) and 3-year-old (PN3) P. notoginseng plant root samples. Principal component analysis of metabolome and transcriptome data revealed major differences between the two groups (PN2 vs. PN3). A total of 1813 metabolites and 28,587 genes were detected in this study, of which 255 metabolites and 3141 genes were found to be differential (p < 0.05) between PN2 vs. PN3, respectively. Among differential metabolites and genes, 155 metabolites and 1217 genes were up-regulated, while 100 metabolites and 1924 genes were down-regulated. The KEGG pathway analysis revealed differentially enriched metabolites belonging to class lipids ("13S-hydroperoxy-9Z, 11E-octadecadionic acid", "9S-hydroxy-10E, 12Z-octadecadionic acid", "9S-oxo-10E, 12Z-octadecadionic acid", and "9,10,13-trihydroxy-11-octadecadionic acid"), nucleotides and derivatives (guanine and cytidine), and phenolic acids (chlorogenic acid) were found to be enriched (p < 0.05) in PN3 compared to PN2. Further, these differentially enriched metabolites were found to be significantly (p < 0.05) regulated via linoleic acid metabolism, nucleotide metabolism, plant hormone signal transduction, and arachidonic acid metabolism pathways. Furthermore, the transcriptome analysis showed the up-regulation of key genes MAT, DMAS, SDH, gallate 1-beta-glucosyltransferase, and beta-D-glucosidase in various plants' secondary metabolic pathways and SAUR, GID1, PP2C, ETR, CTR1, EBF1/2, and ERF1/2 genes observed in phytohormone signal transduction pathway that is involved in plant growth and development, and protection against the various stressors. This study concluded that the roots of a 3-year-old P. notoginseng plant have better metabolome and transcriptome profiles compared to a 2-year-old plant with importantly enriched metabolites and genes in pathways related to metabolism, plant hormone signal transduction, and various biological processes. These findings provide insights into the plant's dynamic biochemical and molecular changes during its growth that have several implications regarding its therapeutic use.
Collapse
Affiliation(s)
- Xinru Yan
- Yunnan Provincial Key Laboratory for Conservation and Utilization of In-Forest Resource, International Ecological Forestry Research Center of Kunming, Southwest Forestry University, Kunming 650224, China; (X.Y.); (A.Z.); (J.J.)
| | - Ao Zhang
- Yunnan Provincial Key Laboratory for Conservation and Utilization of In-Forest Resource, International Ecological Forestry Research Center of Kunming, Southwest Forestry University, Kunming 650224, China; (X.Y.); (A.Z.); (J.J.)
| | - Yiming Guan
- Institute of Special Wild Economic Animal and Plant Science, Chinese Academy of Agricultural Sciences, Changchun 130112, China;
| | - Jinlong Jiao
- Yunnan Provincial Key Laboratory for Conservation and Utilization of In-Forest Resource, International Ecological Forestry Research Center of Kunming, Southwest Forestry University, Kunming 650224, China; (X.Y.); (A.Z.); (J.J.)
| | - Murad Ghanim
- Department of Entomology, Institute of Plant Protection, 68 Hamaccabim Road, Rishon LeZion 7505101, Israel;
| | - Yayu Zhang
- Institute of Special Wild Economic Animal and Plant Science, Chinese Academy of Agricultural Sciences, Changchun 130112, China;
| | - Xiahong He
- Yunnan Provincial Key Laboratory for Conservation and Utilization of In-Forest Resource, International Ecological Forestry Research Center of Kunming, Southwest Forestry University, Kunming 650224, China; (X.Y.); (A.Z.); (J.J.)
| | - Rui Shi
- Yunnan Provincial Key Laboratory for Conservation and Utilization of In-Forest Resource, International Ecological Forestry Research Center of Kunming, Southwest Forestry University, Kunming 650224, China; (X.Y.); (A.Z.); (J.J.)
| |
Collapse
|
43
|
Zhang XY, Xia KR, Wang YN, Liu P, Shang EX, Liu CY, Liu YP, Qu D, Li WW, Duan JA, Chen Y, Zhang HQ. Unraveling the pharmacodynamic substances and possible mechanism of Trichosanthis Pericarpium in the treatment of coronary heart disease based on plasma pharmacochemistry, network pharmacology and experimental validation. JOURNAL OF ETHNOPHARMACOLOGY 2024; 325:117869. [PMID: 38342153 DOI: 10.1016/j.jep.2024.117869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 01/29/2024] [Accepted: 02/04/2024] [Indexed: 02/13/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Coronary heart disease (CHD) is a chronic disease that seriously threatens people's health and even their lives. Currently, there is no ideal drug without side effects for the treatment of CHD. Trichosanthis Pericarpium (TP) has been used for several years in the treatment of diseases associated with CHD. However, there is still a need for systematic research to unravel the pharmacodynamic substances and possible mechanism of TP in the treatment of coronary heart. AIM OF THE STUDY The purpose of current study was to explore the pharmacodynamic substances and potential mechanisms of TP in the treatment of CHD via integrating network pharmacology with plasma pharmacochemistry and experimental validation. MATERIALS AND METHODS The effect of TP intervention in CHD was firstly assessed on high-fat diet combined with isoprenaline-induced CHD rats and H2O2-induced H9c2 cells, respectively. Then, the LC-MS was utilized to identify the absorbed components of TP in the plasma of CHD rats, and this was used to develop a network pharmacology prediction to obtain the possible active components and mechanisms of action. Molecular docking and immunohistochemistry were used to explore the interaction between TP and key targets. Subsequently, the efficacy of the active ingredients was investigated by in vitro cellular experiments, and their metabolic pathways in CHD rats were further analyzed. RESULTS The effects of TP on amelioration of CHD were verified by in vivo and in vitro experiments. Plasma pharmacochemistry and network pharmacology screened six active components in plasma including apigenin, phenylalanine, quercetin, linoleic acid, luteolin, and tangeretin. The interaction of these compounds with potential key targets AKT1, IL-1β, IL-6, TNF-α and VEGFA were preliminarily verified by molecular docking. And immunohistochemical results showed that TP reduced the expression of AKT1, IL-1β, IL-6, TNF-α and VEGFA in CHD rat hearts. Then cellular experiments confirmed that apigenin, phenylalanine, quercetin, linoleic acid, luteolin, and tangeretin were able to reduce the ROS level in H2O2-induced HUVEC cells and promote the migration and tubule formation of HUVEC cells, indicating the pharmacodynamic effects of the active components. Meanwhile, the metabolites of TP in CHD rats suggested that the pharmacological effects of TP might be the result of the combined effects of the active ingredients and their metabolites. CONCLUSION Our study found that TP intervention in CHD is characterized by multi-component and multi-target regulation. Apigenin, phenylalanine, linoleic acid, quercetin, luteolin, and tangeretin are the main active components of TP. TP could reduce inflammatory response and endothelial damage by regulating AKT1, IL-1β, IL-6, TNF-α and VEGFA, reduce ROS level to alleviate the oxidative stress situation and improve heart disease by promoting angiogenesis to regulate endothelial function. This study also provides an experimental and scientific basis for the clinical application and rational development of TP.
Collapse
Affiliation(s)
- Xiao-Yu Zhang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210028, China
| | - Kai-Rou Xia
- Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Ya-Ni Wang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210028, China
| | - Pei Liu
- Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Er-Xin Shang
- Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Cong-Yan Liu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210028, China
| | - Yu-Ping Liu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210028, China
| | - Ding Qu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210028, China
| | - Wei-Wen Li
- Institute of Horticulture, Anhui Academy of Agricultural Sciences, Hefei, 230031, China
| | - Jin-Ao Duan
- Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yan Chen
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210028, China.
| | - Huang-Qin Zhang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210028, China.
| |
Collapse
|
44
|
Zhang F, Zhang Y, Zhou Q, Shi Y, Gao X, Zhai S, Zhang H. Using machine learning to identify proteomic and metabolomic signatures of stroke in atrial fibrillation. Comput Biol Med 2024; 173:108375. [PMID: 38569232 DOI: 10.1016/j.compbiomed.2024.108375] [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: 12/28/2023] [Revised: 03/18/2024] [Accepted: 03/24/2024] [Indexed: 04/05/2024]
Abstract
Atrial fibrillation (AF) is a common cardiac arrhythmia, with stroke being its most detrimental comorbidity. The exact mechanism of AF related stroke (AFS) still needs to be explored. In this study, we integrated proteomics and metabolomics platform to explore disordered plasma proteins and metabolites between AF patients and AFS patients. There were 22 up-regulated and 31 down-regulated differentially expressed proteins (DEPs) in AFS plasma samples. Moreover, 63 up-regulated and 51 down-regulated differentially expressed metabolites (DEMs) were discovered in AFS plasma samples. We integrated proteomics and metabolomics based on the topological interactions of DEPs and DEMs, which yielded revealed several related pathways such as arachidonic acid metabolism, serotonergic synapse, purine metabolism, tyrosine metabolism and steroid hormone biosynthesis. We then performed a machine learning model to identify potential biomarkers of stroke in AF. Finally, we selected 6 proteins and 6 metabolites as candidate biomarkers for predicting stroke in AF by random forest, the area under the curve being 0.976. In conclusion, this study provides new perspectives for understanding the progressive mechanisms of AF related stroke and discovering innovative biomarkers for determining the prognosis of stroke in AF.
Collapse
Affiliation(s)
- Fan Zhang
- NHC Key Laboratory of Cell Transplantation, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Ying Zhang
- Beidahuang Industry Group General Hospital, Harbin, 150001, China
| | - Qi Zhou
- Research Management Office, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Yuanqi Shi
- Key Laboratory of Cardiovascular Disease Acousto-Optic Electromagnetic Diagnosis and Treatment in Heilongjiang Province, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Xiangyuan Gao
- NHC Key Laboratory of Cell Transplantation, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Siqi Zhai
- NHC Key Laboratory of Cell Transplantation, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Haiyu Zhang
- Key Laboratory of Cardiovascular Disease Acousto-Optic Electromagnetic Diagnosis and Treatment in Heilongjiang Province, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China.
| |
Collapse
|
45
|
Zhou J, Hou HT, Song Y, Zhou XL, Chen HX, Zhang LL, Xue HM, Yang Q, He GW. Metabolomics Analysis Identifies Differential Metabolites as Biomarkers for Acute Myocardial Infarction. Biomolecules 2024; 14:532. [PMID: 38785939 PMCID: PMC11117998 DOI: 10.3390/biom14050532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 04/07/2024] [Accepted: 04/23/2024] [Indexed: 05/25/2024] Open
Abstract
Myocardial infarction (MI), including ST-segment elevation MI (STEMI) and non-ST-segment elevation MI (NSTEMI), is still a leading cause of death worldwide. Metabolomics technology was used to explore differential metabolites (DMs) as potential biomarkers for early diagnosis of STEMI and NSTEMI. In the study, 2531 metabolites, including 1925 DMs, were discovered. In the selected 27 DMs, 14 were successfully verified in a new cohort, and the AUC values were all above 0.8. There were 10 in STEMI group, namely L-aspartic acid, L-acetylcarnitine, acetylglycine, decanoylcarnitine, hydroxyphenyllactic acid, ferulic acid, itaconic acid, lauroylcarnitine, myristoylcarnitine, and cis-4-hydroxy-D-proline, and 5 in NSTEMI group, namely L-aspartic acid, arachidonic acid, palmitoleic acid, D-aspartic acid, and palmitelaidic acid. These 14 DMs may be developed as biomarkers for the early diagnosis of MI with high sensitivity and specificity. These findings have particularly important clinical significance for NSTEMI patients because these patients have no typical ECG changes.
Collapse
Affiliation(s)
- Jie Zhou
- Department of Cardiac Surgery & The Institute of Cardiovascular Diseases, TEDA International Cardiovascular Hospital, Tianjin University, Tianjin 300457, China; (J.Z.); (H.-T.H.); (H.-X.C.); (L.-L.Z.); (H.-M.X.); (Q.Y.)
- Tianjin Key Laboratory of Molecular Regulation of Cardiovascular Diseases and Translational Medicine, Tianjin 300457, China; (Y.S.); (X.-L.Z.)
| | - Hai-Tao Hou
- Department of Cardiac Surgery & The Institute of Cardiovascular Diseases, TEDA International Cardiovascular Hospital, Tianjin University, Tianjin 300457, China; (J.Z.); (H.-T.H.); (H.-X.C.); (L.-L.Z.); (H.-M.X.); (Q.Y.)
- Tianjin Key Laboratory of Molecular Regulation of Cardiovascular Diseases and Translational Medicine, Tianjin 300457, China; (Y.S.); (X.-L.Z.)
- Department of Cardiac Surgery & The Institute of Cardiovascular Diseases, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences, Tianjin 300457, China
| | - Yu Song
- Tianjin Key Laboratory of Molecular Regulation of Cardiovascular Diseases and Translational Medicine, Tianjin 300457, China; (Y.S.); (X.-L.Z.)
- Department of Cardiology & The Institute of Cardiovascular Diseases and the Critical Care Unit, TEDA International Cardiovascular Hospital, Tianjin University, Chinese Academy of Medical Sciences, Tianjin 300457, China
| | - Xiao-Lin Zhou
- Tianjin Key Laboratory of Molecular Regulation of Cardiovascular Diseases and Translational Medicine, Tianjin 300457, China; (Y.S.); (X.-L.Z.)
- Department of Cardiology & The Institute of Cardiovascular Diseases and the Critical Care Unit, TEDA International Cardiovascular Hospital, Tianjin University, Chinese Academy of Medical Sciences, Tianjin 300457, China
| | - Huan-Xin Chen
- Department of Cardiac Surgery & The Institute of Cardiovascular Diseases, TEDA International Cardiovascular Hospital, Tianjin University, Tianjin 300457, China; (J.Z.); (H.-T.H.); (H.-X.C.); (L.-L.Z.); (H.-M.X.); (Q.Y.)
- Tianjin Key Laboratory of Molecular Regulation of Cardiovascular Diseases and Translational Medicine, Tianjin 300457, China; (Y.S.); (X.-L.Z.)
- Department of Cardiac Surgery & The Institute of Cardiovascular Diseases, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences, Tianjin 300457, China
| | - Li-Li Zhang
- Department of Cardiac Surgery & The Institute of Cardiovascular Diseases, TEDA International Cardiovascular Hospital, Tianjin University, Tianjin 300457, China; (J.Z.); (H.-T.H.); (H.-X.C.); (L.-L.Z.); (H.-M.X.); (Q.Y.)
- Tianjin Key Laboratory of Molecular Regulation of Cardiovascular Diseases and Translational Medicine, Tianjin 300457, China; (Y.S.); (X.-L.Z.)
- Department of Cardiac Surgery & The Institute of Cardiovascular Diseases, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences, Tianjin 300457, China
| | - Hong-Mei Xue
- Department of Cardiac Surgery & The Institute of Cardiovascular Diseases, TEDA International Cardiovascular Hospital, Tianjin University, Tianjin 300457, China; (J.Z.); (H.-T.H.); (H.-X.C.); (L.-L.Z.); (H.-M.X.); (Q.Y.)
- Tianjin Key Laboratory of Molecular Regulation of Cardiovascular Diseases and Translational Medicine, Tianjin 300457, China; (Y.S.); (X.-L.Z.)
- Department of Cardiac Surgery & The Institute of Cardiovascular Diseases, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences, Tianjin 300457, China
| | - Qin Yang
- Department of Cardiac Surgery & The Institute of Cardiovascular Diseases, TEDA International Cardiovascular Hospital, Tianjin University, Tianjin 300457, China; (J.Z.); (H.-T.H.); (H.-X.C.); (L.-L.Z.); (H.-M.X.); (Q.Y.)
- Tianjin Key Laboratory of Molecular Regulation of Cardiovascular Diseases and Translational Medicine, Tianjin 300457, China; (Y.S.); (X.-L.Z.)
- Department of Cardiac Surgery & The Institute of Cardiovascular Diseases, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences, Tianjin 300457, China
| | - Guo-Wei He
- Department of Cardiac Surgery & The Institute of Cardiovascular Diseases, TEDA International Cardiovascular Hospital, Tianjin University, Tianjin 300457, China; (J.Z.); (H.-T.H.); (H.-X.C.); (L.-L.Z.); (H.-M.X.); (Q.Y.)
- Tianjin Key Laboratory of Molecular Regulation of Cardiovascular Diseases and Translational Medicine, Tianjin 300457, China; (Y.S.); (X.-L.Z.)
- Department of Cardiac Surgery & The Institute of Cardiovascular Diseases, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences, Tianjin 300457, China
| |
Collapse
|
46
|
Li H, Wang L, Zhang H, Yu W, Li Y, Jiang H, Wang D, Wang Y. Study on material basis and anti-hypertensive metabolomics of Zhengan-Xifeng-Tang(ZXT): A comparison between ZXT decoction and granules. J Chromatogr B Analyt Technol Biomed Life Sci 2024; 1236:124063. [PMID: 38447242 DOI: 10.1016/j.jchromb.2024.124063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 01/24/2024] [Accepted: 02/19/2024] [Indexed: 03/08/2024]
Abstract
High blood pressure is a serious human health problem and one of the leading risk factors for fatal complications in cardiovascular disease. The ZXT granules were prepared based on the Zhengan-Xifeng-Tang (ZXT) decoction. However, the therapeutic effects of ZXT granules on spontaneous hypertension and the metabolic pathways in which they may intervene are unclear. The aim of this study was to investigate the antihypertensive effect of ZXT granules on spontaneously hypertensive rats (SHR) and to analyze the metabolic pathway of intervention through chemical composition characterization, pharmacodynamics, and serum metabolomics analysis. After eight weeks of administration, serum and aortic arch samples were collected for biochemical, histopathology and serum metabolomics analysis to assess the effect of ZXT granules on SHR. The results showed that ZXT granules reduced aortic arch injury and blood pressure in SHR rats. Serum data from rats in each group was collected using LC-MS and 74 potential biomarkers were identified that showed significant differences between the model and control groups. Of these, 18 potential biomarkers were found to be deregulated after intervention with ZXT granules. These 18 potential differential metabolic markers are primarily involved in bile acid biosynthesis, arachidonic acid metabolism pathway, and fatty acid degradation. The results demonstrated that ZXT granules significantly affected blood lipids, aortic arch, and metabolic disorders in SHR rats. ZXT granules offer a new possibility for effective and convenient treatment of hypertensive patients.
Collapse
Affiliation(s)
- Haichao Li
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Lihua Wang
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Hao Zhang
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Wenchi Yu
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Yunlun Li
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Haiqing Jiang
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China; Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan 250355, China; Shandong Provincial Key Laboratory of Traditional Chinese Medicine for Basic Research, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Danyang Wang
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China; Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan 250355, China; Shandong Provincial Key Laboratory of Traditional Chinese Medicine for Basic Research, Shandong University of Traditional Chinese Medicine, Jinan 250355, China.
| | - Yu Wang
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250011, China.
| |
Collapse
|
47
|
Panchali T, Dutta A, Das P, Khatun A, Kar R, Mondal S, Mondal KC, Chakrabarti S, Ghosh K, Pradhan S. Amelioration of obesity induction by a high-fat diet and related inflammation by Phasa fish (Setipinna phasa) oil in BALB/c mice. J Appl Biomed 2024; 22:49-58. [PMID: 38505970 DOI: 10.32725/jab.2024.003] [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: 12/24/2022] [Accepted: 01/31/2024] [Indexed: 03/21/2024] Open
Abstract
We have extracted and characterized Phasa fish (Setipinna phasa) oil for the first time to evaluate the anti-obesity and related anti-inflammatory effects on obese mice. Inbred male albino BALB/c mice were segregated into three categories: control (C), Obese control group (OC), and Phasa fish oil treated group (TX). To establish the potentiality of Setipinna phasa oil for its anti-obesity and anti-inflammatory properties, it was extracted and characterized using GC-MS method. To evaluate the anti-obesity effect, different parameters were considered, such as body weight, lipid composition, obesity, and obesity associated inflammation. The physicochemical characteristics of Phasa fish oil revealed that the oil quality was good because acid value, peroxide value, p-anisidine value, Totox value, refractive index, and saponification value were within the standard value range. The GC-MS study explored the presence of fatty acids beneficial to health such as Hexadec-9-enoic acid; Octadec-11-enoic acid; EPA, DHA, Methyl Linolenate, etc. The application of Setipinna phasa oil on the treated mice group acutely lowered body weight and serum lipid profile compared to the obese group. In connection with this, leptin, FAS, and pro-inflammatory cytokines TNF-α genes expression were downregulated in the treated group compared to the obese group. The Phasa oil treated group had an elevated expression of PPAR-α, adiponectin, LPL gene, and anti-inflammatory markers IL-10 and IL-1Ra compared to the obese group. This study suggests that Phasa fish oil, enriched with essential fatty acid, might be used as an anti-obesity and anti-inflammatory supplement.
Collapse
Affiliation(s)
- Titli Panchali
- Midnapore City College, Department of Paramedical and Allied Health Sciences, Bhadutala, Paschim Medinipur, West Bengal, India
| | - Ananya Dutta
- Midnapore City College, Department of Paramedical and Allied Health Sciences, Bhadutala, Paschim Medinipur, West Bengal, India
| | - Pipika Das
- Midnapore City College, Department of Paramedical and Allied Health Sciences, Bhadutala, Paschim Medinipur, West Bengal, India
| | - Amina Khatun
- Midnapore City College, Department of Paramedical and Allied Health Sciences, Bhadutala, Paschim Medinipur, West Bengal, India
| | - Riya Kar
- Midnapore City College, Department of Paramedical and Allied Health Sciences, Bhadutala, Paschim Medinipur, West Bengal, India
| | - Subhadeep Mondal
- Midnapore City College, Department of Paramedical and Allied Health Sciences, Bhadutala, Paschim Medinipur, West Bengal, India
| | | | - Sudipta Chakrabarti
- Midnapore City College, Department of Paramedical and Allied Health Sciences, Bhadutala, Paschim Medinipur, West Bengal, India
| | - Kuntal Ghosh
- Midnapore City College, Department of Paramedical and Allied Health Sciences, Bhadutala, Paschim Medinipur, West Bengal, India
| | - Shrabani Pradhan
- Midnapore City College, Department of Paramedical and Allied Health Sciences, Bhadutala, Paschim Medinipur, West Bengal, India
| |
Collapse
|
48
|
Shen Q, Yan F, Li YW, Wang J, Ji J, Yan WX, He DC, Song P, Shi TQ. Expansion of YALIcloneHR toolkit for Yarrowia lipolytica combined with Golden Gate and CRISPR technology. Biotechnol Lett 2024; 46:37-46. [PMID: 38064043 DOI: 10.1007/s10529-023-03444-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 09/29/2023] [Accepted: 10/17/2023] [Indexed: 01/14/2024]
Abstract
Metabolic Engineering of yeast is a critical approach to improving the production capacity of cell factories. To obtain genetically stable recombinant strains, the exogenous DNA is preferred to be integrated into the genome. Previously, we developed a Golden Gate toolkit YALIcloneNHEJ, which could be used as an efficient modular cloning toolkit for the random integration of multigene pathways through the innate non-homologous end-joining repair mechanisms of Yarrowia lipolytica. We expanded the toolkit by designing additional building blocks of homologous arms and using CRISPR technology. The reconstructed toolkit was thus entitled YALIcloneHR and designed for gene-specific knockout and integration. To verify the effectiveness of the system, the gene PEX10 was selected as the target for the knockout. This system was subsequently applied for the arachidonic acid production, and the reconstructed strain can accumulate 4.8% of arachidonic acid. The toolkit will expand gene editing technology in Y. lipolytica, which would help produce other chemicals derived from acetyl-CoA in the future.
Collapse
Affiliation(s)
- Qi Shen
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, 2 Xuelin Road, Qixia District, Nanjing, 210023, People's Republic of China
| | - Fang Yan
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, 2 Xuelin Road, Qixia District, Nanjing, 210023, People's Republic of China
| | - Ya-Wen Li
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, 2 Xuelin Road, Qixia District, Nanjing, 210023, People's Republic of China
| | - Jian Wang
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, 2 Xuelin Road, Qixia District, Nanjing, 210023, People's Republic of China
| | - Jia Ji
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, 2 Xuelin Road, Qixia District, Nanjing, 210023, People's Republic of China
| | - Wen-Xin Yan
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, 2 Xuelin Road, Qixia District, Nanjing, 210023, People's Republic of China
| | - Dan-Chen He
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, 2 Xuelin Road, Qixia District, Nanjing, 210023, People's Republic of China
| | - Ping Song
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, 2 Xuelin Road, Qixia District, Nanjing, 210023, People's Republic of China
| | - Tian-Qiong Shi
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, 2 Xuelin Road, Qixia District, Nanjing, 210023, People's Republic of China.
| |
Collapse
|
49
|
Martinez-Morata I, Wu H, Galvez-Fernandez M, Ilievski V, Bottiglieri T, Niedzwiecki MM, Goldsmith J, Jones DP, Kioumourtzoglou MA, Pierce B, Walker DI, Gamble MV. Metabolomic Effects of Folic Acid Supplementation in Adults: Evidence from the FACT Trial. J Nutr 2024; 154:670-679. [PMID: 38092151 PMCID: PMC10900167 DOI: 10.1016/j.tjnut.2023.12.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 12/04/2023] [Accepted: 12/08/2023] [Indexed: 12/31/2023] Open
Abstract
BACKGROUND Folic acid (FA) is the oxidized form of folate found in supplements and FA-fortified foods. Most FA is reduced by dihydrofolate reductase to 5-methyltetrahydrofolate (5mTHF); the latter is the form of folate naturally found in foods. Ingestion of FA increases the plasma levels of both 5mTHF and unmetabolized FA (UMFA). Limited information is available on the downstream metabolic effects of FA supplementation, including potential effects associated with UMFA. OBJECTIVE We aimed to assess the metabolic effects of FA-supplementation, and the associations of plasma 5mTHF and UMFA with the metabolome in FA-naïve Bangladeshi adults. METHODS Sixty participants were selected from the Folic Acid and Creatine Trial; half received 800 μg FA/day for 12 weeks and half placebo. Plasma metabolome profiles were measured by high-resolution mass spectrometry, including 170 identified metabolites and 26,541 metabolic features. Penalized regression methods were used to assess the associations of targeted metabolites with FA-supplementation, plasma 5mTHF, and plasma UMFA. Pathway analyses were conducted using Mummichog. RESULTS In penalized models of identified metabolites, FA-supplementation was associated with higher choline. Changes in 5mTHF concentrations were positively associated with metabolites involved in amino acid metabolism (5-hydroxyindoleacetic acid, acetylmethionine, creatinine, guanidinoacetate, hydroxyproline/n-acetylalanine) and 2 fatty acids (docosahexaenoic acid and linoleic acid). Changes in 5mTHF concentrations were negatively associated with acetylglutamate, acetyllysine, carnitine, propionyl carnitine, cinnamic acid, homogentisate, arachidonic acid, and nicotine. UMFA concentrations were associated with lower levels of arachidonic acid. Together, metabolites selected across all models were related to lipids, aromatic amino acid metabolism, and the urea cycle. Analyses of nontargeted metabolic features identified additional pathways associated with FA supplementation. CONCLUSION In addition to the recapitulation of several expected metabolic changes associated with 5mTHF, we observed additional metabolites/pathways associated with FA-supplementation and UMFA. Further studies are needed to confirm these associations and assess their potential implications for human health. TRIAL REGISTRATION NUMBER This trial was registered at https://clinicaltrials.gov as NCT01050556.
Collapse
Affiliation(s)
- Irene Martinez-Morata
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, United States
| | - Haotian Wu
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, United States
| | - Marta Galvez-Fernandez
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, United States
| | - Vesna Ilievski
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, United States
| | - Teodoro Bottiglieri
- Center of Metabolomics, Institute of Metabolic Disease, Baylor Scott & White Research Institute, Dallas, TX, United States
| | - Megan M Niedzwiecki
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Jeff Goldsmith
- Department of Biostatistics, Mailman School of Public Health, Columbia University, New York, NY, United States
| | - Dean P Jones
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, GA, United States; Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, United States
| | - Marianthi-Anna Kioumourtzoglou
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, United States
| | - Brandon Pierce
- Department of Public Health Sciences, University of Chicago, Chicago, IL, United States; Department of Human Genetics, University of Chicago, Chicago, IL, United States; Comprehensive Cancer Center, University of Chicago, Chicago, IL, United States
| | - Douglas I Walker
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, United States
| | - Mary V Gamble
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, United States.
| |
Collapse
|
50
|
Song J, Luo C, Liu Z, Liu J, Xie L, Zhang X, Xie Z, Li X, Ma Z, Ding J, Li H, Xiang H. Early fecal microbiota transplantation from high abdominal fat chickens affects recipient cecal microbiome and metabolism. Front Microbiol 2024; 14:1332230. [PMID: 38260901 PMCID: PMC10800977 DOI: 10.3389/fmicb.2023.1332230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 12/18/2023] [Indexed: 01/24/2024] Open
Abstract
Abdominal fat deposition (AFD) in chickens is closely related to the gut microecological balance. In this study, the gut microbiota from high-AFD chickens was transplanted into the same strain of 0-day-old chicks via fecal microbiota transplantation (FMT). The FTM from chickens with high AFD had no obvious effects on growth traits, adult body weight, carcass weight, abdominal fat weight, and abdominal fat percentage, but did reduce the coefficient of variation of AFD traits. FMT significantly decreased cecal microbiome richness, changed the microbiota structure, and regulated the biological functions associated with energy metabolism and fat synthesis. Additionally, the cecal metabolite composition and metabolic function of FMT recipient chickens were also significantly altered from those of the controls. Transplantation of high-AFD chicken gut microbiota promoted fatty acid elongation and biosynthesis and reduced the metabolism of vitamins, steroids, and carbohydrates in the cecum. These findings provide insights into the mechanisms by which chicken gut microbiota affect host metabolic profiles and fat deposition.
Collapse
Affiliation(s)
- Jiani Song
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, School of Life Science and Engineering, Foshan University, Foshan, China
| | - Chaowei Luo
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, School of Life Science and Engineering, Foshan University, Foshan, China
| | - Zhijie Liu
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, School of Life Science and Engineering, Foshan University, Foshan, China
| | - Jingshou Liu
- Guangdong Tinoo’s Foods Group Co., Ltd., Guangdong, China
| | - Li Xie
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, School of Life Science and Engineering, Foshan University, Foshan, China
| | - Xing Zhang
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, School of Life Science and Engineering, Foshan University, Foshan, China
| | - Zhuojun Xie
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, School of Life Science and Engineering, Foshan University, Foshan, China
| | - Xiangkun Li
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, School of Life Science and Engineering, Foshan University, Foshan, China
| | - Zheng Ma
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, School of Life Science and Engineering, Foshan University, Foshan, China
| | - Jinlong Ding
- Guangdong Tinoo’s Foods Group Co., Ltd., Guangdong, China
| | - Hua Li
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, School of Life Science and Engineering, Foshan University, Foshan, China
- Guangdong Tinoo’s Foods Group Co., Ltd., Guangdong, China
| | - Hai Xiang
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, School of Life Science and Engineering, Foshan University, Foshan, China
| |
Collapse
|