1
|
Xu Y, Yang Y, Shi Y, Li B, Xie Y, Le G. Dietary methionine supplementation improves cognitive dysfunction associated with transsulfuration pathway upregulation in subacute aging mice. NPJ Sci Food 2024; 8:104. [PMID: 39702349 DOI: 10.1038/s41538-024-00348-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Accepted: 11/25/2024] [Indexed: 12/21/2024] Open
Abstract
To explore the effects of methionine (Met) supplementation on cognitive dysfunction and the associated mechanisms in aging mice. The mice were administrated 0.15 g/kg/day D-galactose subcutaneously and fed a normal (0.86% Met) or a Met-supplemented diet (1.72% Met) for 11 weeks. Behavioral experiments were conducted, and we measured the plasma metabolite levels, hippocampal and plasma redox and inflammatory states, and hippocampal transsulfuration pathway-related parameters. Met supplementation prevented aging-induced anxiety and cognitive deficiencies, and normalized the plasma levels of multiple systemic metabolites (e.g., betaine, taurine, and choline). Furthermore, dietary Met supplementation abolished oxidative stress and inflammation, selectively modulated the expression of multiple cognition-related genes and proteins, and increased flux via the transsulfuration pathway in the hippocampi of aging mice, with significant increase in H2S and glutathione production. Our findings suggest that dietary Met supplementation prevented cognitive deficiencies in aging mice, probably because of increased flux via the transsulfuration pathway.
Collapse
Affiliation(s)
- Yuncong Xu
- Henan Key Laboratory of cereal and Oil Food Safety Inspection and Control, College of Food Science and Engineering, Henan University of Technology, Zhengzhou, Henan, China
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, China
| | - Yuhui Yang
- Henan Key Laboratory of cereal and Oil Food Safety Inspection and Control, College of Food Science and Engineering, Henan University of Technology, Zhengzhou, Henan, China.
| | - Yonghui Shi
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, China
| | - Bowen Li
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, China
| | - Yanli Xie
- Henan Key Laboratory of cereal and Oil Food Safety Inspection and Control, College of Food Science and Engineering, Henan University of Technology, Zhengzhou, Henan, China
| | - Guowei Le
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, China
| |
Collapse
|
2
|
Jia Y, Zhao Y, Niu M, Zhao C, Li X, Chen H. Preliminary study of metabonomic changes during the progression of atherosclerosis in miniature pigs. Animal Model Exp Med 2024; 7:419-432. [PMID: 38923366 PMCID: PMC11369038 DOI: 10.1002/ame2.12462] [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: 09/26/2023] [Accepted: 05/19/2024] [Indexed: 06/28/2024] Open
Abstract
BACKGROUND To explore potential biomarkers for early diagnosis of atherosclerosis (AS) and provide basic data for further research on AS, the characteristics of serum metabolomics during the progression of AS in mini-pigs were observed dynamically. METHODS An AS model in Bama miniature pigs was established by a high-cholesterol and high-fat diet. Fasting serum samples were collected monthly for metabolomics and serum lipid detection. At the end of the treatment period, pathological analysis of the abdominal aorta and coronary artery was performed to evaluate the lesions of AS, thereby distinguishing the susceptibility of mini-pigs to AS. The metabolomics was detected using a high-resolution untargeted metabolomic approach. Statistical analysis was used to identify metabolites associated with AS susceptibility. RESULTS Based on pathological analysis, mini-pigs were divided into two groups: a susceptible group (n = 3) and a non-susceptible group (n = 6). A total of 1318 metabolites were identified, with significant shifting of metabolic profiles over time in both groups. Dynamic monitoring analysis highlighted 57 metabolites that exhibited an obvious trend of differential changes between two groups with the advance of time. The KEGG (Kyoto Encyclopedia of Genes and Genomes) pathway enrichment analysis indicated significant disorders in cholesterol metabolism, primary bile acid metabolism, histidine metabolism, as well as taurine and hypotaurine metabolism. CONCLUSIONS During the progression of AS in mini-pigs induced by high-cholesterol/high-fat diet, the alterations in serum metabolic profile exhibited a time-dependent pattern, accompanied by notable disturbances in lipid metabolism, cholesterol metabolism, and amino acid metabolism. These metabolites may become potential biomarkers for early diagnosis of AS.
Collapse
Affiliation(s)
- Yunxiao Jia
- Laboratory Animal CenterChinese PLA General HospitalBeijingPeople's Republic of China
| | - Yuqiong Zhao
- Laboratory Animal CenterChinese PLA General HospitalBeijingPeople's Republic of China
| | - Miaomiao Niu
- Laboratory Animal CenterChinese PLA General HospitalBeijingPeople's Republic of China
| | - Changqi Zhao
- Laboratory Animal CenterChinese PLA General HospitalBeijingPeople's Republic of China
| | - Xuezhuang Li
- Laboratory Animal CenterChinese PLA General HospitalBeijingPeople's Republic of China
| | - Hua Chen
- Laboratory Animal CenterChinese PLA General HospitalBeijingPeople's Republic of China
| |
Collapse
|
3
|
Liu P, Chen Q, Zhang L, Ren C, Shi B, Zhang J, Wang S, Chen Z, Wang Q, Xie H, Huang Q, Tang H. Rapid quantification of 50 fatty acids in small amounts of biological samples for population molecular phenotyping. BIOPHYSICS REPORTS 2023; 9:299-308. [PMID: 38524698 PMCID: PMC10960574 DOI: 10.52601/bpr.2023.230042] [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: 11/25/2023] [Accepted: 12/15/2023] [Indexed: 03/26/2024] Open
Abstract
Efficient quantification of fatty-acid (FA) composition (fatty-acidome) in biological samples is crucial for understanding physiology and pathophysiology in large population cohorts. Here, we report a rapid GC-FID/MS method for simultaneous quantification of all FAs in numerous biological matrices. Within eight minutes, this method enabled simultaneous quantification of 50 FAs as fatty-acid methyl esters (FAMEs) in femtomole levels following the efficient transformation of FAs in all lipids including FFAs, cholesterol-esters, glycerides, phospholipids and sphingolipids. The method showed satisfactory inter-day and intra-day precision, stability and linearity (R2 > 0.994) within a concentration range of 2-3 orders of magnitude. FAs were then quantified in typical multiple biological matrices including human biofluids (urine, plasma) and cells, animal intestinal content and tissue samples. We also established a quantitative structure-retention relationship (QSRR) for analytes to accurately predict their retention time and aid their reliable identification. We further developed a novel no-additive retention index (NARI) with endogenous FAMEs reducing inter-batch variations to 15 seconds; such NARI performed better than the alkanes-based classical RI, making meta-analysis possible for data obtained from different batches and platforms. Collectively, this provides an inexpensive high-throughput analytical system for quantitative phenotyping of all FAs in 8-minutes multiple biological matrices in large cohort studies of pathophysiological effects.
Collapse
Affiliation(s)
- Pinghui Liu
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Metabonomics and Systems Biology Laboratory at Shanghai International Centre for Molecular Phenomics, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Qinsheng Chen
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Metabonomics and Systems Biology Laboratory at Shanghai International Centre for Molecular Phenomics, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Lianglong Zhang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Metabonomics and Systems Biology Laboratory at Shanghai International Centre for Molecular Phenomics, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Chengcheng Ren
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Metabonomics and Systems Biology Laboratory at Shanghai International Centre for Molecular Phenomics, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Biru Shi
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Metabonomics and Systems Biology Laboratory at Shanghai International Centre for Molecular Phenomics, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Jingxian Zhang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Metabonomics and Systems Biology Laboratory at Shanghai International Centre for Molecular Phenomics, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Shuaiyao Wang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Metabonomics and Systems Biology Laboratory at Shanghai International Centre for Molecular Phenomics, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Ziliang Chen
- Wuhan Laboratory for Shanghai Metabolome Institute (SMI) Ltd, Wuhan 430000, China
| | - Qi Wang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Metabonomics and Systems Biology Laboratory at Shanghai International Centre for Molecular Phenomics, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Hui Xie
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Metabonomics and Systems Biology Laboratory at Shanghai International Centre for Molecular Phenomics, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Qingxia Huang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Metabonomics and Systems Biology Laboratory at Shanghai International Centre for Molecular Phenomics, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Huiru Tang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Metabonomics and Systems Biology Laboratory at Shanghai International Centre for Molecular Phenomics, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| |
Collapse
|
4
|
Du Z, Li F, Jiang L, Li L, Du Y, Yu H, Luo Y, Wang Y, Sun H, Hu C, Li J, Yang Y, Jiao X, Wang L, Qin Y. Metabolic systems approaches update molecular insights of clinical phenotypes and cardiovascular risk in patients with homozygous familial hypercholesterolemia. BMC Med 2023; 21:275. [PMID: 37501168 PMCID: PMC10375787 DOI: 10.1186/s12916-023-02967-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 06/28/2023] [Indexed: 07/29/2023] Open
Abstract
BACKGROUND Homozygous familial hypercholesterolemia (HoFH) is an orphan metabolic disease characterized by extremely elevated low-density lipoprotein cholesterol (LDL-C), xanthomas, aortic stenosis, and premature atherosclerotic cardiovascular disease (ASCVD). In addition to LDL-C, studies in experimental models and small clinical populations have suggested that other types of metabolic molecules might also be risk factors responsible for cardiovascular complications in HoFH, but definitive evidence from large-scale human studies is still lacking. Herein, we aimed to comprehensively characterize the metabolic features and risk factors of human HoFH by using metabolic systems strategies. METHODS Two independent multi-center cohorts with a total of 868 individuals were included in the cross-sectional study. First, comprehensive serum metabolome/lipidome-wide analyses were employed to identify the metabolomic patterns for differentiating HoFH patients (n = 184) from heterozygous FH (HeFH, n = 376) and non-FH (n = 100) subjects in the discovery cohort. Then, the metabolomic patterns were verified in the validation cohort with 48 HoFH patients, 110 HeFH patients, and 50 non-FH individuals. Subsequently, correlation/regression analyses were performed to investigate the associations of clinical/metabolic alterations with typical phenotypes of HoFH. In the prospective study, a total of 84 HoFH patients with available follow-up were enrolled from the discovery cohort. Targeted metabolomics, deep proteomics, and random forest approaches were performed to investigate the ASCVD-associated biomarkers in HoFH patients. RESULTS Beyond LDL-C, various bioactive metabolites in multiple pathways were discovered and validated for differentiating HoFH from HoFH and non-FH. Our results demonstrated that the inflammation and oxidative stress-related metabolites in the pathways of arachidonic acid and lipoprotein(a) metabolism were independently associated with the prevalence of corneal arcus, xanthomas, and supravalvular/valvular aortic stenosis in HoFH patients. Our results also identified a small marker panel consisting of high-density lipoprotein cholesterol, lipoprotein(a), apolipoprotein A1, and eight proinflammatory and proatherogenic metabolites in the pathways of arachidonic acid, phospholipid, carnitine, and sphingolipid metabolism that exhibited significant performances on predicting first ASCVD events in HoFH patients. CONCLUSIONS Our findings demonstrate that human HoFH is associated with a variety of metabolic abnormalities and is more complex than previously known. Furthermore, this study provides additional metabolic alterations that hold promise as residual risk factors in HoFH population.
Collapse
Affiliation(s)
- Zhiyong Du
- Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, National Clinical Research Center for Cardiovascular Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China
- Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing, 100029, China
| | - Fan Li
- Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, National Clinical Research Center for Cardiovascular Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China
- Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing, 100029, China
| | - Long Jiang
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi Province, China
| | - Linyi Li
- Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, National Clinical Research Center for Cardiovascular Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China
- Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing, 100029, China
| | - Yunhui Du
- Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, National Clinical Research Center for Cardiovascular Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China
- Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing, 100029, China
| | - Huahui Yu
- Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, National Clinical Research Center for Cardiovascular Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China
- Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing, 100029, China
| | - Yan Luo
- Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, National Clinical Research Center for Cardiovascular Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China
- Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing, 100029, China
| | - Yu Wang
- Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, National Clinical Research Center for Cardiovascular Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China
- Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing, 100029, China
| | - Haili Sun
- Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, National Clinical Research Center for Cardiovascular Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China
- Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing, 100029, China
| | - Chaowei Hu
- Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, National Clinical Research Center for Cardiovascular Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China
- Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing, 100029, China
| | - Jianping Li
- Department of Cardiology, Peking University First Hospital, Beijing, 100034, China
| | - Ya Yang
- Suzhou Municipal Hospital, Suzhou, 215002, Jiangsu Province, China
| | - Xiaolu Jiao
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Department of Cardiology, College of Medicine, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, 310020, Zhejiang Province, China
| | - Luya Wang
- Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, National Clinical Research Center for Cardiovascular Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China.
- Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing, 100029, China.
| | - Yanwen Qin
- Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, National Clinical Research Center for Cardiovascular Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China.
- Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing, 100029, China.
| |
Collapse
|
5
|
Lipidized PrRP Analog Exhibits Strong Anti-Obesity and Antidiabetic Properties in Old WKY Rats with Obesity and Glucose Intolerance. Nutrients 2023; 15:nu15020280. [PMID: 36678151 PMCID: PMC9864151 DOI: 10.3390/nu15020280] [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: 11/25/2022] [Revised: 12/28/2022] [Accepted: 12/30/2022] [Indexed: 01/08/2023] Open
Abstract
Prolactin-releasing peptide (PrRP) is an anorexigenic neuropeptide that has potential for the treatment of obesity and its complications. Recently, we designed a palmitoylated PrRP31 analog (palm11-PrRP31) that is more stable than the natural peptide and able to act centrally after peripheral administration. This analog acted as an anti-obesity and glucose-lowering agent, attenuating lipogenesis in rats and mice with high-fat (HF) diet-induced obesity. In Wistar Kyoto (WKY) rats fed a HF diet for 52 weeks, we explored glucose intolerance, but also prediabetes, liver steatosis and insulin resistance-related changes, as well as neuroinflammation in the brain. A potential beneficial effect of 6 weeks of treatment with palm11-PrRP31 and liraglutide as comparator was investigated. Liver lipid profiles, as well as urinary and plasma metabolomic profiles, were measured by lipidomics and metabolomics, respectively. Old obese WKY rats showed robust glucose intolerance that was attenuated by palm11-PrRP31, but not by liraglutide treatment. On the contrary, liraglutide had a beneficial effect on insulin resistance parameters. Despite obesity and prediabetes, WKY rats did not develop steatosis owing to HF diet feeding, even though liver lipogenesis was enhanced. Plasma triglycerides and cholesterol were not increased by HFD feeding, which points to unincreased lipid transport from the liver. The liver lipid profile was significantly altered by a HF diet that remained unaffected by palm11-PrRP31 or liraglutide treatment. The HF-diet-fed WKY rats revealed astrogliosis in the brain cortex and hippocampus, which was attenuated by treatment. In conclusion, this study suggested multiple beneficial anti-obesity-related effects of palm11-PrRP31 and liraglutide in both the periphery and brain.
Collapse
|
6
|
Sakanaka A, Katakami N, Furuno M, Nishizawa H, Omori K, Taya N, Ishikawa A, Mayumi S, Inoue M, Tanaka Isomura E, Amano A, Shimomura I, Fukusaki E, Kuboniwa M. Salivary metabolic signatures of carotid atherosclerosis in patients with type 2 diabetes hospitalized for treatment. Front Mol Biosci 2022; 9:1074285. [PMID: 36619162 PMCID: PMC9815705 DOI: 10.3389/fmolb.2022.1074285] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 11/28/2022] [Indexed: 12/24/2022] Open
Abstract
Atherosclerosis is a life-threatening disease associated with morbidity and mortality in patients with type 2 diabetes (T2D). This study aimed to characterize a salivary signature of atherosclerosis based on evaluation of carotid intima-media thickness (IMT) to develop a non-invasive predictive tool for diagnosis and disease follow-up. Metabolites in saliva and plasma samples collected at admission and after treatment from 25 T2D patients hospitalized for 2 weeks to undergo medical treatment for diabetes were comprehensively profiled using metabolomic profiling with gas chromatography-mass spectrometry. Orthogonal partial least squares analysis, used to explore the relationships of IMT with clinical markers and plasma and salivary metabolites, showed that the top predictors for IMT included salivary allantoin and 1,5-anhydroglucitol (1,5-AG) at both the baseline examination at admission and after treatment. Furthermore, though treatment induced alterations in salivary levels of allantoin and 1,5-AG, it did not modify the association between IMT and these metabolites (p interaction > 0.05), and models with these metabolites combined yielded satisfactory diagnostic accuracy for the high IMT group even after treatment (area under curve = 0.819). Collectively, this salivary metabolite combination may be useful for non-invasive identification of T2D patients with a higher atherosclerotic burden in clinical settings.
Collapse
Affiliation(s)
- Akito Sakanaka
- Department of Preventive Dentistry, Osaka University Graduate School of Dentistry, Suita, Japan
| | - Naoto Katakami
- Department of Metabolic Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - Masahiro Furuno
- Department of Biotechnology, Osaka University Graduate School of Engineering, Suita, Japan
| | - Hitoshi Nishizawa
- Department of Metabolic Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - Kazuo Omori
- Department of Metabolic Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - Naohiro Taya
- Department of Metabolic Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - Asuka Ishikawa
- Department of Preventive Dentistry, Osaka University Graduate School of Dentistry, Suita, Japan
| | - Shota Mayumi
- Department of Preventive Dentistry, Osaka University Graduate School of Dentistry, Suita, Japan
| | - Moe Inoue
- Department of Preventive Dentistry, Osaka University Graduate School of Dentistry, Suita, Japan
| | - Emiko Tanaka Isomura
- First Department of Oral and Maxillofacial Surgery, Osaka University Graduate School of Dentistry, Suita, Japan
| | - Atsuo Amano
- Department of Preventive Dentistry, Osaka University Graduate School of Dentistry, Suita, Japan
| | - Iichiro Shimomura
- Department of Metabolic Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - Eiichiro Fukusaki
- Department of Biotechnology, Osaka University Graduate School of Engineering, Suita, Japan
| | - Masae Kuboniwa
- Department of Preventive Dentistry, Osaka University Graduate School of Dentistry, Suita, Japan,*Correspondence: Masae Kuboniwa,
| |
Collapse
|
7
|
Du Z, Li F, Li L, Wang Y, Li J, Yang Y, Jiang L, Wang L, Qin Y. Low-density lipoprotein receptor genotypes modify the sera metabolome of patients with homozygous familial hypercholesterolemia. iScience 2022; 25:105334. [DOI: 10.1016/j.isci.2022.105334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 08/30/2022] [Accepted: 10/10/2022] [Indexed: 10/31/2022] Open
|
8
|
High dietary methionine intake may contribute to the risk of nonalcoholic fatty liver disease by inhibiting hepatic H2S production. Food Res Int 2022; 158:111507. [DOI: 10.1016/j.foodres.2022.111507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 06/09/2022] [Accepted: 06/10/2022] [Indexed: 12/06/2022]
|
9
|
Wang Y, Xu Y, Xu X, Wang H, Wang D, Yan W, Zhu J, Hao H, Wang G, Cao L, Zhang J. Ginkgo biloba extract ameliorates atherosclerosis via rebalancing gut flora and microbial metabolism. Phytother Res 2022; 36:2463-2480. [PMID: 35312112 DOI: 10.1002/ptr.7439] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 02/17/2022] [Accepted: 02/24/2022] [Indexed: 12/31/2022]
Abstract
The Ginkgo biloba leave extract (GbE) is widely applied in the prevention and treatment of atherosclerotic cardiovascular diseases in clinical practice. However, its mechanism of actions has not been totally elucidated. In this study, we confirmed the beneficial effects of GbE in alleviating hypercholesterolemia, inflammation and atherosclerosis in Ldlr-/- mice, which were fed 12 weeks of Western diet (WD). Moreover, 16S rRNA sequencing revealed that GbE treatment reshaped the WD-perturbed intestinal microbiota, particularly decreased the Firmicutes/Bacteroidetes ratio and elevated the abundance of Akkermansia, Alloprevotella, Alistipes, and Parabacteroides. Furthermore, GbE treatment downregulated the intestinal transcriptional levels of proinflammatory cytokines and enhanced the expression of tight junction proteins, exerting the roles of attenuating the intestinal inflammation as well as repairing the gut barrier. Meanwhile, the targeted metabolomic analysis displayed that GbE treatment significantly reversed the dysfunction of the microbial metabolic phenotypes, including promoting the production of short chain fatty acids, indole-3-acetate and secondary bile acids, which were correlated with the atherosclerotic plaque areas. Finally, we confirmed GbE-altered gut microbiota was sufficient to alleviate atherosclerosis by fecal microbiota transplantation. In summary, our findings provide important insights into the pharmacological mechanism underlying the antiatherogenic efficacy of GbE.
Collapse
Affiliation(s)
- Yun Wang
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, China
| | - Yuanyuan Xu
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, China
| | - Xiaowei Xu
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, China
| | - Hong Wang
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, China
| | - Dong Wang
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, China
| | - Wenchao Yan
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, China
| | - Jiaying Zhu
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, China
| | - Haiping Hao
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, China
| | - Guangji Wang
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, China
| | - Lijuan Cao
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, China
| | - Jun Zhang
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, China.,School of Pharmacy, Nanjing Medical University, Nanjing, China
| |
Collapse
|
10
|
Hu SH, He XD, Nie J, Hou JL, Wu J, Liu XY, Wei Y, Tang HR, Sun WX, Zhou SX, Yuan YY, An YP, Yan GQ, Lin Y, Lin PC, Zhao JJ, Ye ML, Zhao JY, Xu W, Zhao SM. Methylene-bridge tryptophan fatty acylation regulates PI3K-AKT signaling and glucose uptake. Cell Rep 2022; 38:110509. [PMID: 35294873 DOI: 10.1016/j.celrep.2022.110509] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 09/15/2021] [Accepted: 02/16/2022] [Indexed: 12/01/2022] Open
Abstract
Protein fatty acylation regulates numerous cell signaling pathways. Polyunsaturated fatty acids (PUFAs) exert a plethora of physiological effects, including cell signaling regulation, with underlying mechanisms to be fully understood. Herein, we report that docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA) regulate PI3K-AKT signaling by modifying PDK1 and AKT2. DHA-administered mice exhibit altered phosphorylation of proteins in signaling pathways. Methylene bridge-containing DHA/EPA acylate δ1 carbon of tryptophan 448/543 in PDK1 and tryptophan 414 in AKT2 via free radical pathway, recruit both the proteins to the cytoplasmic membrane, and activate PI3K signaling and glucose uptake in a tryptophan acylation-dependent but insulin-independent manner in cultured cells and in mice. DHA/EPA deplete cytosolic PDK1 and AKT2 and induce insulin resistance. Akt2 knockout in mice abrogates DHA/EPA-induced PI3K-AKT signaling. Our results identify PUFA's methylene bridge tryptophan acylation, a protein fatty acylation that regulates cell signaling and may underlie multifaceted effects of methylene-bridge-containing PUFAs.
Collapse
Affiliation(s)
- Song-Hua Hu
- Obstetrics & Gynecology Hospital of Fudan University, Institutes of Metabolism and Integrative Biology, State Key Laboratory of Genetic Engineering, School of Life Sciences and Institutes of Biomedical Sciences, Shanghai 200438, P.R. China; NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Shanghai Key Laboratory of Medical Epigenetics, and Children's Hospital of Fudan University, Shanghai 200438, P.R. China
| | - Xia-Di He
- Obstetrics & Gynecology Hospital of Fudan University, Institutes of Metabolism and Integrative Biology, State Key Laboratory of Genetic Engineering, School of Life Sciences and Institutes of Biomedical Sciences, Shanghai 200438, P.R. China; NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Shanghai Key Laboratory of Medical Epigenetics, and Children's Hospital of Fudan University, Shanghai 200438, P.R. China
| | - Ji Nie
- Obstetrics & Gynecology Hospital of Fudan University, Institutes of Metabolism and Integrative Biology, State Key Laboratory of Genetic Engineering, School of Life Sciences and Institutes of Biomedical Sciences, Shanghai 200438, P.R. China; NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Shanghai Key Laboratory of Medical Epigenetics, and Children's Hospital of Fudan University, Shanghai 200438, P.R. China
| | - Jun-Li Hou
- Department of Chemistry, Fudan University, Shanghai 200438, P.R. China
| | - Jiang Wu
- Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei 230027, P. R. China
| | - Xiao-Yan Liu
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, National Chromatographic R&A Center, Dalian Institute of Chemical Physics, Chinese Academy of Sciences (CAS), Dalian 116023, China
| | - Yun Wei
- Obstetrics & Gynecology Hospital of Fudan University, Institutes of Metabolism and Integrative Biology, State Key Laboratory of Genetic Engineering, School of Life Sciences and Institutes of Biomedical Sciences, Shanghai 200438, P.R. China; NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Shanghai Key Laboratory of Medical Epigenetics, and Children's Hospital of Fudan University, Shanghai 200438, P.R. China
| | - Hui-Ru Tang
- Obstetrics & Gynecology Hospital of Fudan University, Institutes of Metabolism and Integrative Biology, State Key Laboratory of Genetic Engineering, School of Life Sciences and Institutes of Biomedical Sciences, Shanghai 200438, P.R. China
| | - Wen-Xing Sun
- Obstetrics & Gynecology Hospital of Fudan University, Institutes of Metabolism and Integrative Biology, State Key Laboratory of Genetic Engineering, School of Life Sciences and Institutes of Biomedical Sciences, Shanghai 200438, P.R. China; NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Shanghai Key Laboratory of Medical Epigenetics, and Children's Hospital of Fudan University, Shanghai 200438, P.R. China
| | - Shu-Xian Zhou
- Obstetrics & Gynecology Hospital of Fudan University, Institutes of Metabolism and Integrative Biology, State Key Laboratory of Genetic Engineering, School of Life Sciences and Institutes of Biomedical Sciences, Shanghai 200438, P.R. China; NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Shanghai Key Laboratory of Medical Epigenetics, and Children's Hospital of Fudan University, Shanghai 200438, P.R. China
| | - Yi-Yuan Yuan
- Obstetrics & Gynecology Hospital of Fudan University, Institutes of Metabolism and Integrative Biology, State Key Laboratory of Genetic Engineering, School of Life Sciences and Institutes of Biomedical Sciences, Shanghai 200438, P.R. China; NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Shanghai Key Laboratory of Medical Epigenetics, and Children's Hospital of Fudan University, Shanghai 200438, P.R. China
| | - Yan-Peng An
- Obstetrics & Gynecology Hospital of Fudan University, Institutes of Metabolism and Integrative Biology, State Key Laboratory of Genetic Engineering, School of Life Sciences and Institutes of Biomedical Sciences, Shanghai 200438, P.R. China
| | - Guo-Quan Yan
- Obstetrics & Gynecology Hospital of Fudan University, Institutes of Metabolism and Integrative Biology, State Key Laboratory of Genetic Engineering, School of Life Sciences and Institutes of Biomedical Sciences, Shanghai 200438, P.R. China
| | - Yan Lin
- Obstetrics & Gynecology Hospital of Fudan University, Institutes of Metabolism and Integrative Biology, State Key Laboratory of Genetic Engineering, School of Life Sciences and Institutes of Biomedical Sciences, Shanghai 200438, P.R. China; NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Shanghai Key Laboratory of Medical Epigenetics, and Children's Hospital of Fudan University, Shanghai 200438, P.R. China
| | - Peng-Cheng Lin
- Key Laboratory for Tibet Plateau Phytochemistry of Qinghai Province, College of Pharmacy, Qinghai University for Nationalities, Xining 810007, P. R. China
| | - Jean J Zhao
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA; Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Ming-Liang Ye
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, National Chromatographic R&A Center, Dalian Institute of Chemical Physics, Chinese Academy of Sciences (CAS), Dalian 116023, China.
| | - Jian-Yuan Zhao
- Obstetrics & Gynecology Hospital of Fudan University, Institutes of Metabolism and Integrative Biology, State Key Laboratory of Genetic Engineering, School of Life Sciences and Institutes of Biomedical Sciences, Shanghai 200438, P.R. China; NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Shanghai Key Laboratory of Medical Epigenetics, and Children's Hospital of Fudan University, Shanghai 200438, P.R. China.
| | - Wei Xu
- Obstetrics & Gynecology Hospital of Fudan University, Institutes of Metabolism and Integrative Biology, State Key Laboratory of Genetic Engineering, School of Life Sciences and Institutes of Biomedical Sciences, Shanghai 200438, P.R. China; NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Shanghai Key Laboratory of Medical Epigenetics, and Children's Hospital of Fudan University, Shanghai 200438, P.R. China.
| | - Shi-Min Zhao
- Obstetrics & Gynecology Hospital of Fudan University, Institutes of Metabolism and Integrative Biology, State Key Laboratory of Genetic Engineering, School of Life Sciences and Institutes of Biomedical Sciences, Shanghai 200438, P.R. China; NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Shanghai Key Laboratory of Medical Epigenetics, and Children's Hospital of Fudan University, Shanghai 200438, P.R. China; Key Laboratory for Tibet Plateau Phytochemistry of Qinghai Province, College of Pharmacy, Qinghai University for Nationalities, Xining 810007, P. R. China.
| |
Collapse
|
11
|
Yang H, Li H, Chen W, Mei Z, Yuan Y, Wang X, Chu L, Xu Y, Sun Y, Li D, Gao H, Zhan B, Li H, Yang X. Therapeutic Effect of Schistosoma japonicum Cystatin on Atherosclerotic Renal Damage. Front Cell Dev Biol 2021; 9:760980. [PMID: 34901005 PMCID: PMC8656285 DOI: 10.3389/fcell.2021.760980] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 10/11/2021] [Indexed: 01/15/2023] Open
Abstract
Atherosclerosis is a chronic inflammation of the arterial vessel wall driven by lipid metabolism disorders. Although helminthic infection and their derivatives have been identified to attenuate the chronic inflammatory diseases, the immunomodulatory effect of recombinant Schistosoma japonicum cystatin (rSj-Cys) on metabolic diseases and atherosclerosis has not been reported. In this study, we investigated the therapeutic efficacy of rSj-Cys on atherosclerotic renal damage and explored the related immunological mechanism. The results demonstrated that treatment with rSj-Cys significantly reduced body weight gain, hyperlipidemia, and atherosclerosis induced by the high-fat diet in apoE–/– mice. The treatment of rSj-Cys also significantly improved kidney functions through promoting macrophage polarization from M1 to M2, therefore inhibiting M1 macrophage–induced inflammation. The possible mechanism underlying the regulatory effect of rSj-Cys on reducing atherosclerosis and atherosclerotic renal damage is that rSj-Cys stimulates regulatory T cell and M2 macrophage polarization that produce regulatory cytokines, such as interleukin 10 and transforming growth factor β. The therapeutic effect of rSj-Cys on atherosclerotic renal damage is possibly through inhibiting the activation of TLR2/Myd88 signaling pathway. The results in this study provide evidence for the first time that Schistosoma-derived cystatin could be developed as a therapeutic agent to treat lipid metabolism disorder and atherosclerosis that threats million lives around the world.
Collapse
Affiliation(s)
- Huijuan Yang
- Department of Nephrology, First Affiliated Hospital of Bengbu Medical College, Bengbu, China.,Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, China
| | - Hongqi Li
- Department of Gerontology, Anhui Provincial Hospital, First Affiliated Hospital of University of Science and Technology of China, Hefei, China
| | - Weidong Chen
- Department of Nephrology, First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Zhijie Mei
- Department of Urology, First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Yuan Yuan
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, China.,Basic Medical College of Bengbu Medical College, Bengbu, China
| | - Xiaoli Wang
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, China.,Basic Medical College of Bengbu Medical College, Bengbu, China
| | - Liang Chu
- Second Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Yu Xu
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, China.,Basic Medical College of Bengbu Medical College, Bengbu, China
| | - Yan Sun
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, China.,Basic Medical College of Bengbu Medical College, Bengbu, China
| | - Dingru Li
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, China.,Basic Medical College of Bengbu Medical College, Bengbu, China
| | - Hongyu Gao
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, China.,Basic Medical College of Bengbu Medical College, Bengbu, China
| | - Bin Zhan
- National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, United States
| | - Huihui Li
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, China.,Basic Medical College of Bengbu Medical College, Bengbu, China
| | - Xiaodi Yang
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, China.,Basic Medical College of Bengbu Medical College, Bengbu, China
| |
Collapse
|
12
|
Yang Y, Qian J, Li B, Lu M, Le G, Xie Y. Metabolomics Based on 1H-NMR Reveal the Regulatory Mechanisms of Dietary Methionine Restriction on Splenic Metabolic Dysfunction in Obese Mice. Foods 2021; 10:foods10102439. [PMID: 34681487 PMCID: PMC8535630 DOI: 10.3390/foods10102439] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Revised: 10/06/2021] [Accepted: 10/12/2021] [Indexed: 12/13/2022] Open
Abstract
Methionine restriction (MR) has been reported to have many beneficial health effects, including stress resistance enhancement and lifespan extension. However, the effects of MR on the splenic metabolic dysfunction induced by obesity in mice remain unknown. This study aimed to investigate the scientific problem and clarify its possible mechanisms. C57BL/6J mice in the control group were fed a control diet (0.86% methionine, 4.2% fat) for 34 weeks, and others were fed a high-fat diet (0.86% methionine, 24% fat) for 10 weeks to establish diet-induced obese (DIO) mouse models. Then, the obtained DIO mice were randomly divided into two groups: the DIO group (DIO diet), the DIO + MR group (0.17% methionine, 24% fat) for 24 weeks. Our results indicated that MR decreased spleen weight, and spleen and plasma lipid profiles, promoted lipid catabolism and fatty acid oxidation, glycolysis and tricarboxylic acid cycle metabolism, and improved mitochondrial function and ATP generation in the spleen. Moreover, MR normalized the splenic redox state and inflammation-related metabolite levels, and increased plasma levels of immunoglobulins. Furthermore, MR increased percent lean mass and splenic crude protein levels, activated the autophagy pathway and elevated nucleotide synthesis to maintain protein synthesis in the spleen. These findings indicate that MR can ameliorate metabolic dysfunction by reducing lipid accumulation, oxidative stress, and inflammation in the spleen, and the mechanism may be the activation of autophagy pathway.
Collapse
Affiliation(s)
- Yuhui Yang
- College of Food Science and Engineering, Henan University of Technology, Zhengzhou 450001, China; (Y.Y.); (J.Q.); (M.L.)
| | - Jing Qian
- College of Food Science and Engineering, Henan University of Technology, Zhengzhou 450001, China; (Y.Y.); (J.Q.); (M.L.)
| | - Bowen Li
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi 214122, China; (B.L.); (G.L.)
| | - Manman Lu
- College of Food Science and Engineering, Henan University of Technology, Zhengzhou 450001, China; (Y.Y.); (J.Q.); (M.L.)
| | - Guowei Le
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi 214122, China; (B.L.); (G.L.)
| | - Yanli Xie
- College of Food Science and Engineering, Henan University of Technology, Zhengzhou 450001, China; (Y.Y.); (J.Q.); (M.L.)
- Correspondence: ; Tel.: +86-371-6775-8022
| |
Collapse
|
13
|
Zhang L, Ma X, Liu P, Ge W, Hu L, Zuo Z, Xiao H, Liao W. Treatment and mechanism of fecal microbiota transplantation in mice with experimentally induced ulcerative colitis. Exp Biol Med (Maywood) 2021; 246:1563-1575. [PMID: 33926254 DOI: 10.1177/15353702211006044] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Restoring intestinal microbiota dysbiosis with fecal microbiota transplantation is considered as a promising treatment for ulcerative colitis. However, the mechanisms underlying its relieving effects remain unclear. Ulcerative colitis pathogenesis is associated with the involvement of immune cells and inflammatory cytokines. Here, we aimed to investigate the effect of fecal microbiota transplantation on T cell cytokines in a dextran sulfate sodium-induced ulcerative colitis mouse model. Five-aminosalicylic acid (5-ASA) was used as the positive control. Male C57BL/6 mice were randomly assigned to control, model (UC), UC + FMT, and UC + 5-ASA groups. Each group consisted of five mice. The establishment of the mouse model was verified by fecal occult-blood screening and hematoxylin-eosin staining. Results showed that fecal microbiota transplantation reduced colonic inflammation, significantly decreased T helper (Th)1 and Th17 cells, interferon-gamma, interleukin-2 and interleukin-17, as well as significantly increased Th2 and regulatory T (Treg) cells, interleukin-4, interleukin-10, and transforming growth factor-beta, and improved routine blood count. Furthermore, 16S rRNA gene-sequencing analysis showed a significant increase in the relative abundance of genus Akkermansia and a significant decrease in the relative abundance of genus Helicobacter in the ulcerative colitis group. Fecal microbiota transplantation restored the profile of the intestinal microbiota to that of the control group. These findings demonstrated the capability of fecal microbiota transplantation in controlling experimentally induced ulcerative colitis by improving Th1/Th2 and Th17/Treg imbalance through the regulation of intestinal microbiota.
Collapse
Affiliation(s)
- Leichang Zhang
- Department of Anorectal, The Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang 330006, P.R. China
| | - Xiaofei Ma
- Department of Anorectal, Department of Postgraduate Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, P.R. China
| | - Peng Liu
- Department of Anorectal, Department of Postgraduate Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, P.R. China
| | - Wei Ge
- Department of Anorectal, The Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang 330006, P.R. China
| | - Lixia Hu
- Department of Traditional Chinese Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, P.R. China
| | - Zhengyun Zuo
- Formula-Pattern Research Center, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, P.R. China
| | - Huirong Xiao
- Department of Anorectal, The Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang 330006, P.R. China
| | - Wu Liao
- Department of Anorectal, The Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang 330006, P.R. China
| |
Collapse
|
14
|
Deng L, Fu D, Zhu L, Huang J, Ling Y, Cai Z. Testosterone deficiency accelerates early stage atherosclerosis in miniature pigs fed a high-fat and high-cholesterol diet: urine 1H NMR metabolomics targeted analysis. Mol Cell Biochem 2020; 476:1245-1255. [PMID: 33226572 DOI: 10.1007/s11010-020-03987-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 11/16/2020] [Indexed: 01/17/2023]
Abstract
To gain insights into the role of testosterone in the development of atherosclerosis and its related metabolic pathways, we applied a proton nuclear magnetic resonance (1H NMR)-based metabolomics approach to investigate urine metabolic profiles in miniature pigs fed a high-fat and high-cholesterol (HFC) diet among intact male pigs (IM), castrated male pigs (CM) and castrated male pigs with testosterone replacement (CMT). Our results showed that testosterone deficiency significantly increased atherosclerotic lesion areas, intima-media thickness, as well as serum lipid levels in the CM pigs. Moreover, seventeen significantly changed metabolites were identified in both IM vs. CM and CMT vs. CM groups. Among these, seven were shared between the two comparative groups and were all significantly reduced in the urine of the CM group but rescued in the CMT group. In addition, the correlation analysis demonstrated that several metabolites, including niacinamide, myo-inositol, choline and 3-hydroxyisovalerate, were negatively correlated with atherosclerotic lesion areas. Our study demonstrated that testosterone deficiency accelerated early AS formation in HFC diet-fed pigs, which involved several metabolites predominantly related to lipid metabolism, inflammation, oxidative stress and endothelial disorders. Our results reveal potential pathways in the pathogenesis of atherosclerosis caused by testosterone deficiency and HFC diet.
Collapse
Affiliation(s)
- Liqun Deng
- Laboratory Animal Research Center, Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China.,Institute of Comparative Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Danting Fu
- Laboratory Animal Research Center, Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China.,Department of Experimental Animals, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou, 310012, China
| | - Liang Zhu
- Laboratory Animal Research Center, Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China.,Institute of Comparative Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Junjie Huang
- Laboratory Animal Research Center, Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China.,Institute of Comparative Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Yun Ling
- Laboratory Animal Research Center, Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China.,Institute of Comparative Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Zhaowei Cai
- Laboratory Animal Research Center, Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China. .,Institute of Comparative Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| |
Collapse
|
15
|
Geng J, Xu H, Fu W, Yu X, Xu G, Cao H, Lin G, Sui D. Rosuvastatin protects against endothelial cell apoptosis in vitro and alleviates atherosclerosis in ApoE -/- mice by suppressing endoplasmic reticulum stress. Exp Ther Med 2020; 20:550-560. [PMID: 32537013 PMCID: PMC7282009 DOI: 10.3892/etm.2020.8733] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 04/01/2020] [Indexed: 12/11/2022] Open
Abstract
The development of abnormal lipid-induced atherosclerosis is initiated with endothelial cell apoptosis. Vascular endothelial cells possess highly developed endoplasmic reticulum (ER), which is involved in lipid metabolism, indicating that ER stress may contribute chiefly to the induction of endothelial cell apoptosis. Based on its ability to reduce cholesterol levels, rosuvastatin may play an endothelial and vascular protective role by regulating ER stress. In the present study, the involvement of the inhibition of the ER stress-induced endothelial injury was investigated in combination with the lipid lowering effects of rosuvastatin. This compound can be used to inhibit cholesterol synthesis in atherosclerosis. Rosuvastatin decreased the apoptotic rates of human umbilical vascular endothelial cells (HUVECs) that had been stimulated with ox-low density lipoprotein (LDL) in vitro and repressed the mRNA levels of CHOP, sXBP1 and caspase-12, and decreased caspase-12 activity, as well as the content of glucose-regulated protein 78 (GRP78), phosphorylated (p)-protein kinase RNA-like ER kinase (PERK), p-inositol-requiring protein 1α (IRE1α) and p-eIF2α proteins. In addition, ApoE-/- mice were fed with atherogenic chow for 8 weeks for atherosclerosis induction and rosuvastatin was provided by intragastric administration for an additional 4 weeks. Subsequently, the atherosclerotic plaque formation in the aorta was evaluated by Oil Red O and hematoxylin and eosin staining, and the serum LDL, high-density lipoprotein, total cholesterol (TC) and triacylglycerol (TG) levels were measured. In addition, the induction of apoptosis of endothelial cells and the expression levels of GRP78, p-PERK, p-IRE1α and p-eIF2α were assessed in the aorta. Rosuvastatin repressed atherosclerotic plaque formation and endothelial apoptosis in the aorta and decreased LDL and TG levels in the serum, as determined by in vivo results. Furthermore, it downregulated the expression levels of protein chaperone GRP78, p-PERK, p-IRE1α and p-eIF2α in the aortic intima. The data indicated that rosuvastatin could protect HUVECs from ER stress-induced apoptosis triggered by oxidized LDL. It could also inhibit atherosclerosis formation in ApoE-/- mice aorta by regulating the PERK/eIF2α/C/EBPα-homologous protein and IRE1α/sXBP1 signaling pathways. Taken collectively, the present study demonstrated the preventive and therapeutic effects of rosuvastatin in protecting from the development of endothelial cell dysfunction diseases.
Collapse
Affiliation(s)
- Jianan Geng
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Huali Xu
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Wenwen Fu
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Xiaofeng Yu
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Guoliang Xu
- Department of Cardiovascular Medicine, the Eastern Division of First Hospital, Jilin University, Changchun, Jilin 130031, P.R. China
| | - Hongyan Cao
- Department of Cardiovascular Medicine, the Eastern Division of First Hospital, Jilin University, Changchun, Jilin 130031, P.R. China
| | - Guangzhu Lin
- Department of Cardiovascular Medicine, the Eastern Division of First Hospital, Jilin University, Changchun, Jilin 130031, P.R. China
| | - Dayun Sui
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
16
|
Hyperlipidemia Affects Tight Junctions and Pump Function in the Corneal Endothelium. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 190:563-576. [PMID: 31945314 DOI: 10.1016/j.ajpath.2019.11.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Revised: 09/29/2019] [Accepted: 11/14/2019] [Indexed: 12/20/2022]
Abstract
Hyperlipidemia impacts on various diseases, such as atherosclerosis, hypertension, and diabetes mellitus. However, its influence, if any, on ocular tissues is largely unknown. Herein, we developed hyperlipidemic murine models by feeding 4-week-old male wild-type mice with a high-fat diet and apolipoprotein E knockout (ApoE-/-) mice with a high-fat diet or standard diet to investigate the corneal endothelial change under hyperlipidemic conditions. Oil Red O staining showed an accumulation of lipid droplets in corneal endothelial cells (CECs) of hyperlipidemic mice. Other manifestations included a reduced cell density and distorted cell morphology, a disruption of the endothelial cell tight junctions and adhesion junctions, a reduced number of surface microvilli, down-regulation of Na+-K+-ATPase expression and function, activation of oxidative stress, changes in mitochondrial ultrastructure, and increased apoptosis. CEC recovery after injury, moreover, was diminished in hyperlipidemic mice; and high palmitate levels were found in the aqueous humor. In vitro hyperlipemia model, moreover, was found to be associated with dose-dependent CEC cytotoxicity, altered cell morphology, reduced pump function, and an induction of oxidative stress, leading to functional and pathologic changes in the corneal endothelium.
Collapse
|
17
|
Ma N, Yang Y, Liu X, Li S, Qin Z, Li J. Plasma metabonomics and proteomics studies on the anti-thrombosis mechanism of aspirin eugenol ester in rat tail thrombosis model. J Proteomics 2019; 215:103631. [PMID: 31891783 DOI: 10.1016/j.jprot.2019.103631] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 12/10/2019] [Accepted: 12/27/2019] [Indexed: 01/09/2023]
Abstract
Aspirin eugenol eater (AEE), a new drug compound, was synthesized through the combination of aspirin and eugenol. Antithrombotic effects of AEE have been confirmed in carrageenan-induced rat tail thrombosis model. However, its mechanism is unclear. With the application of integrated approach combining proteomics and metabolomics, the profilings of protein and metabolite in plasma were examined in thrombosis rat pretreated with AEE, aspirin and eugenol, respectively. A clear separation of the plasma metabolic profiles from different groups was found in score plots. 15 metabolites related with the metabolism of fatty acid, energy and amino acid were found. A total of 144, 38, 41 and 54 differentially abundant proteins (DAPs) were identified in control, AEE, aspirin and eugenol group, respectively. Proteomic results showed that aspirin modulated 7 proteins in amino acid metabolism and 4 proteins in complement system; eugenol regulated the 8 proteins related with coagulation cascades and fibrinogen; AEE improved 3 proteins in TCA cycle and 3 in lipid metabolism. Integrated analysis suggested that AEE improved fatty acid, energy and lipid metabolism to against thrombosis. Results of this study indicated AEE had different action mechanism on thrombosis from aspirin and eugenol, and contribute to understanding the mechanisms of AEE on thrombosis. SIGNIFICANCE: Thrombosis is a threat to human health, and there is an urgent need for new drug. In this study, compared with the model group, plasma metabolic profiles in AEE-treated rats were clearly separated; 15 metabolites and 38 proteins were picked out. These metabolites and proteins may assist in understanding the action mechanism of AEE on thrombosis. The results of plasma metabonomics and proteomics also revealed the different action mechanism among AEE, aspirin and eugenol on thrombosis. This study established the foundation to further evaluate the druggability of AEE on thrombosis treatment.
Collapse
Affiliation(s)
- Ning Ma
- Key Lab of New Animal Drug Project of Gansu Province, Key Lab of Veterinary Pharmaceutical Development, Ministry of Agriculture, Lanzhou Institute of Husbandry and Pharmaceutical Science of Chinese Academy of Agricultural Sciences, Lanzhou 730050, PR China; College of Veterinary Medicine, Agricultural University of Hebei, Baoding, Hebei 071000, PR China
| | - Yajun Yang
- Key Lab of New Animal Drug Project of Gansu Province, Key Lab of Veterinary Pharmaceutical Development, Ministry of Agriculture, Lanzhou Institute of Husbandry and Pharmaceutical Science of Chinese Academy of Agricultural Sciences, Lanzhou 730050, PR China
| | - Xiwang Liu
- Key Lab of New Animal Drug Project of Gansu Province, Key Lab of Veterinary Pharmaceutical Development, Ministry of Agriculture, Lanzhou Institute of Husbandry and Pharmaceutical Science of Chinese Academy of Agricultural Sciences, Lanzhou 730050, PR China
| | - Shihong Li
- Key Lab of New Animal Drug Project of Gansu Province, Key Lab of Veterinary Pharmaceutical Development, Ministry of Agriculture, Lanzhou Institute of Husbandry and Pharmaceutical Science of Chinese Academy of Agricultural Sciences, Lanzhou 730050, PR China
| | - Zhe Qin
- Key Lab of New Animal Drug Project of Gansu Province, Key Lab of Veterinary Pharmaceutical Development, Ministry of Agriculture, Lanzhou Institute of Husbandry and Pharmaceutical Science of Chinese Academy of Agricultural Sciences, Lanzhou 730050, PR China
| | - Jianyong Li
- Key Lab of New Animal Drug Project of Gansu Province, Key Lab of Veterinary Pharmaceutical Development, Ministry of Agriculture, Lanzhou Institute of Husbandry and Pharmaceutical Science of Chinese Academy of Agricultural Sciences, Lanzhou 730050, PR China.
| |
Collapse
|
18
|
Development of a Predictive Model to Induce Atherogenesis and Hepato-Renal Impairment in Female Rats. Biomolecules 2019; 9:biom9110664. [PMID: 31671756 PMCID: PMC6921007 DOI: 10.3390/biom9110664] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 09/17/2019] [Accepted: 09/20/2019] [Indexed: 02/07/2023] Open
Abstract
Therapeutic approaches for the treatment of dyslipidemia and atherosclerosis have radically changed in recent decades. Part of this advance undeniably stems from basic biomedical research that has provided a better understanding and identification of new therapeutic targets. The aim of this work was to develop a model to induce atherogenesis and hepato-renal impairment in female Wistar rats. The following groups received the respective treatments for 60 days: control animals, non-ovariectomized rats that received an atherogenic diet (NEAD), ovariectomized rats that received an atherogenic diet (NOAD), non-ovariectomized rats that received an atherogenic diet and oral Nω-nitro-l-arginine methyl ester hydrochloride (l-NAME; LEAD), and ovariectomized rats that received an atherogenic diet and oral l-NAME (LOAD). Animals in the NEAD, NOAD, LEAD, and LOAD groups also received methimazole and cholecalciferol daily. Urinary, biochemical, hemodynamic, and electrocardiographic parameters and renal function were assessed. Samples of the liver, heart, kidney, and arteries were collected to investigate redox status and perform histopathological analyses. All of the groups developed dyslipidemia and hepatic steatosis. Only the NEAD group developed arterial lesions that were compatible with fatty streaks. Renal function was significantly impaired in the LEAD and NOAD groups. These results indicate a viable alternative to induce atherogenesis and hepato-renal impairment in female rats.
Collapse
|
19
|
Peron G, Sut S, Dal Ben S, Voinovich D, Dall'Acqua S. Untargeted UPLC-MS metabolomics reveals multiple changes of urine composition in healthy adult volunteers after consumption of curcuma longa L. extract. Food Res Int 2019; 127:108730. [PMID: 31882111 DOI: 10.1016/j.foodres.2019.108730] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 09/06/2019] [Accepted: 09/28/2019] [Indexed: 01/21/2023]
Abstract
Curcuma longa L. is used as food supplement to prevent diseases, although limited studies have been performed on healthy subjects up to now. In the present work, an untargeted UPLC-MS metabolomics approach was applied to study the changes of 24-hours urinary composition on healthy volunteers due to a 28-days daily consumption of a dried C. longa extract containing a standardized amount of curcuminoids. Changes in the excretion of different metabolites were observed after supplementation. Curcumin and two metabolic derivatives (hexahydrocurcumin and dihydrocurcumin) were detected in urine, indicating the absorption of the main curcuminoid from the extract and its further metabolism by liver and gut microbiota. For the first time ar-turmerone, the main apolar constituent of curcuma, was detected in urine in intact form, and its presence was confirmed by a targeted GC-MS analysis. The increase of tetranor-PGJM and tetranor-PGDM, two prostaglandin-D2 metabolites, was observed, being related to the anti-inflammatory effect exerted by curcuma. The variation of the amounts of HPAG, PAG, proline-betaine and hydroxyphenyllactic acid indicate that the supplementation induced changes to the activity of gut microbiota. Finally, the reduced excretion of niacin metabolites (nicotinuric acid, trigonelline and 2PY) and medium- and short-chain acylcarnitines suggests that curcuma could induce the mitochondrial β-oxidation of fatty acids for energy production in healthy subjects. Overall, the results indicate that a prolonged daily consumption of a dried curcuma extract exerts multiple effects on healthy subjects, furthermore they show the opportunity offered by untargeted metabolomics for the study of the bioactivity of natural extracts in healthy human volunteers.
Collapse
Affiliation(s)
- Gregorio Peron
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via Marzolo 5, 35131 Padova, Italy.
| | - Stefania Sut
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via Marzolo 5, 35131 Padova, Italy.
| | - Simone Dal Ben
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, Piazzale Europa 1, I-34127 Trieste, Italy
| | - Dario Voinovich
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, Piazzale Europa 1, I-34127 Trieste, Italy.
| | - Stefano Dall'Acqua
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via Marzolo 5, 35131 Padova, Italy.
| |
Collapse
|
20
|
Dong M, Xu X, Huang Q, Lei H, Xu G, Ma J, Hatzakis E, Wang X, Zhang L. Dose-Dependent Effects of Triclocarban Exposure on Lipid Homeostasis in Rats. Chem Res Toxicol 2019; 32:2320-2328. [PMID: 31576746 DOI: 10.1021/acs.chemrestox.9b00316] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Manyuan Dong
- CAS Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Centre for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences (CAS), Wuhan National Research Center for Optoelectronics, Wuhan 430071, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Xiaoyi Xu
- CAS Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Centre for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences (CAS), Wuhan National Research Center for Optoelectronics, Wuhan 430071, P. R. China
- College of Chemistry and Materials Science, South-Central University for Nationalities, Wuhan 430074, P. R. China
| | - Qingxia Huang
- CAS Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Centre for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences (CAS), Wuhan National Research Center for Optoelectronics, Wuhan 430071, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Hehua Lei
- CAS Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Centre for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences (CAS), Wuhan National Research Center for Optoelectronics, Wuhan 430071, P. R. China
| | - Guangyong Xu
- CAS Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Centre for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences (CAS), Wuhan National Research Center for Optoelectronics, Wuhan 430071, P. R. China
- School of Environmental and Safety Engineering, Changzhou University, Jiangsu, 213164, P. R. China
| | - Jianfeng Ma
- School of Environmental and Safety Engineering, Changzhou University, Jiangsu, 213164, P. R. China
| | - Emmanuel Hatzakis
- Department of Food Science and Technology, The Ohio State University, Columbus, Ohio 43210, United States
| | - Xian Wang
- College of Chemistry and Materials Science, South-Central University for Nationalities, Wuhan 430074, P. R. China
| | - Limin Zhang
- CAS Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Centre for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences (CAS), Wuhan National Research Center for Optoelectronics, Wuhan 430071, P. R. China
| |
Collapse
|
21
|
Dey P. Gut microbiota in phytopharmacology: A comprehensive overview of concepts, reciprocal interactions, biotransformations and mode of actions. Pharmacol Res 2019; 147:104367. [PMID: 31344423 DOI: 10.1016/j.phrs.2019.104367] [Citation(s) in RCA: 132] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 07/11/2019] [Accepted: 07/19/2019] [Indexed: 02/07/2023]
Abstract
The dynamic and delicate interactions amongst intestinal microbiota, metabolome and metabolism dictates human health and disease. In recent years, our understanding of gut microbial regulation of intestinal immunometabolic and redox homeostasis have evolved mainly out of in vivo studies associated with high-fat feeding induced metabolic diseases. Techniques utilizing fecal transplantation and germ-free mice have been instrumental in reproducibly demonstrating how the gut microbiota affects disease pathogenesis. However, the pillars of modern drug discovery i.e. evidence-based pharmacological studies critically lack focus on intestinal microflora. This is primarily due to targeted in vitro molecular-approaches at cellular-level that largely overlook the etiology of disease pathogenesis from the physiological perspective. Thus, this review aims to provide a comprehensive understanding of the key notions of intestinal microbiota and dysbiosis, and highlight the microbiota-phytochemical bidirectional interactions that affects bioavailability and bioactivity of parent phytochemicals and their metabolites. Potentially by focusing on the three major aspects of gut microbiota i.e. microbial abundance, diversity, and functions, I will discuss phytochemical-microbiota reciprocal interactions, biotransformation of phytochemicals and plant-derived drugs, and pre-clinical and clinical efficacies of herbal medicine on dysbiosis. Additionally, in relation to phytochemical pharmacology, I will briefly discuss the role of dietary-patterns associated with changes in microbial profiles and review pharmacological study models considering possible microbial effects. This review therefore, emphasize on the timely and critically needed evidence-based phytochemical studies focusing on gut microbiota and will provide newer insights for future pre-clinical and clinical phytopharmacological interventions.
Collapse
Affiliation(s)
- Priyankar Dey
- Human Nutrition Program, Department of Human Sciences, The Ohio State University, Columbus, Ohio, USA.
| |
Collapse
|
22
|
Yan Y, Du Z, Chen C, Li J, Xiong X, Zhang Y, Jiang H. Lysophospholipid profiles of apolipoprotein E-deficient mice reveal potential lipid biomarkers associated with atherosclerosis progression using validated UPLC-QTRAP-MS/MS-based lipidomics approach. J Pharm Biomed Anal 2019; 171:148-157. [PMID: 30999225 DOI: 10.1016/j.jpba.2019.03.062] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 03/25/2019] [Accepted: 03/27/2019] [Indexed: 01/19/2023]
Abstract
Lysophospholipids (Lyso-PLs) are lipid-derived signaling molecules which were demonstrated to have a strong correlation with the progression of atherosclerosis. In this study, we investigated the influence of high-fat diet on Lyso-PL profiles of atherosclerosis-prone apolipoprotein E-deficient (ApoE-/-) mice and wild type C57BL/6 J mice to find out the potential biomarkers associated with atherosclerosis. Firstly, the quantitative profiling method for Lyso-PLs based on ultra-performance liquid chromatography-quadrupole linear ion trap mass spectrometry (UPLC-QTRAP-MS/MS) was established and validated. Secondly, this method was utilized to quantify 169 targeted Lyso-PLs in plasma samples of ApoE-/- mice and wild type C57BL/6 J mice collected at different time points. Finally, 12 of 37 differential Lyso-PLs were identified as more reliable biomarkers by integrating static metabolomics and time-dependent analyses, among which Lyso-PC/15:0, 18:1/Lyso-PI, 22:5/Lyso-PI and 22:4/Lyso-PI were highly correlated with TCand LDL-C levels. Meanwhile, we found that the Lyso-PL profiles of ApoE-/- mice and C57BL/6 J mice were distinguished by altered metabolism of different Lyso-PLs classes, while C57BL/6 J mice fed with high-fat diet and normal diet were discriminated by the content differences of Lyso-PLs with same fatty acid composition. In conclusion, these results provided detailed changes of Lyso-PL profiles associated with atherosclerosis and the differential Lyso-PLs with reasonable change trends may serve as promising biomarkers for atherosclerosis progression.
Collapse
Affiliation(s)
- Yingfei Yan
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan, China
| | - Zhifeng Du
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan, China
| | - Chang Chen
- Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Jiaxin Li
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan, China
| | - Xiang Xiong
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan, China
| | - Yang Zhang
- Department of Pharmacy, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China.
| | - Hongliang Jiang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
23
|
Yang Y, Wang Y, Sun J, Zhang J, Guo H, Shi Y, Cheng X, Tang X, Le G. Dietary methionine restriction reduces hepatic steatosis and oxidative stress in high-fat-fed mice by promoting H2S production. Food Funct 2019; 10:61-77. [DOI: 10.1039/c8fo01629a] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Dietary methionine restriction reduces hepatic steatosis and oxidative stress in high-fat-fed mice by promoting H2S production.
Collapse
Affiliation(s)
- Yuhui Yang
- State Key Laboratory of Food Science and Technology
- Jiangnan University
- Wuxi
- China
- Center for Food Nutrition and Functional Food Engineering
| | - Yanan Wang
- Center for Food Nutrition and Functional Food Engineering
- School of Food Science and Technology
- Jiangnan University
- Wuxi
- China
| | - Jin Sun
- State Key Laboratory of Food Science and Technology
- Jiangnan University
- Wuxi
- China
- Center for Food Nutrition and Functional Food Engineering
| | - Jiahong Zhang
- Center for Food Nutrition and Functional Food Engineering
- School of Food Science and Technology
- Jiangnan University
- Wuxi
- China
| | - Haitao Guo
- Center for Food Nutrition and Functional Food Engineering
- School of Food Science and Technology
- Jiangnan University
- Wuxi
- China
| | - Yonghui Shi
- State Key Laboratory of Food Science and Technology
- Jiangnan University
- Wuxi
- China
- Center for Food Nutrition and Functional Food Engineering
| | - Xiangrong Cheng
- State Key Laboratory of Food Science and Technology
- Jiangnan University
- Wuxi
- China
- Center for Food Nutrition and Functional Food Engineering
| | - Xue Tang
- State Key Laboratory of Food Science and Technology
- Jiangnan University
- Wuxi
- China
- Center for Food Nutrition and Functional Food Engineering
| | - Guowei Le
- State Key Laboratory of Food Science and Technology
- Jiangnan University
- Wuxi
- China
- Center for Food Nutrition and Functional Food Engineering
| |
Collapse
|
24
|
Cognitive impairment correlates with serum carbonyl compound profiles in subclinical carotid atherosclerosis. Neuroreport 2018; 29:1550-1557. [DOI: 10.1097/wnr.0000000000001147] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
25
|
Yang Y, Zhang J, Wu G, Sun J, Wang Y, Guo H, Shi Y, Cheng X, Tang X, Le G. Dietary methionine restriction regulated energy and protein homeostasis by improving thyroid function in high fat diet mice. Food Funct 2018; 9:3718-3731. [PMID: 29978874 DOI: 10.1039/c8fo00685g] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Methionine-restricted diets (MRD) show an integrated series of beneficial health effects, including improving insulin sensitivity, limiting fat deposition, and decreasing oxidative stress, and inflammation responses. We aimed to explore the systemic responses to a MRD in mice fed with a high fat (HFD) and clarify the possible mechanism. Mice were fed with a control diet (0.86% methionine + 4% fat, CON), HFD (0.86% methionine + 20% fat), or MRD (0.17% methionine + 20% fat) for 22 consecutive weeks. HFD-fed mice showed widespread systemic metabolic disorders and thyroid dysfunction. A MRD significantly increased energy expenditure (e.g. fatty acid oxidation, glycolysis, and tricarboxylic acid cycle metabolism), regulated protein homeostasis, improved gut microbiota functions, prevented thyroid dysfunction, increased plasma thyroxine and triiodothyronine levels, decreased plasma thyroid stimulating hormone levels, increased type 2 deiodinase (DIO2) activity, and up-regulated mRNA and protein expression levels of DIO2 and thyroid hormone receptor α1 in the skeletal muscle. These results suggest that a MRD can improve the metabolic disorders induced by a HFD, and especially regulate energy and protein homeostasis likely through improved thyroid function. Thus, reducing methionine intake (e.g. through a vegan diet) may improve metabolic health in animals and humans.
Collapse
Affiliation(s)
- Yuhui Yang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, PR China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Chen J, Zhang C, Wu X, Ji H, Ma W, Wei S, Zhang L, Chen J. 1 H NMR-based nontargeted metabonomics study of plasma and urinary biochemical changes in Kudouzi treated rats. REVISTA BRASILEIRA DE FARMACOGNOSIA 2018. [DOI: 10.1016/j.bjp.2018.05.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
27
|
Deidda M, Noto A, Bassareo PP, Cadeddu Dessalvi C, Mercuro G. Metabolomic Approach to Redox and Nitrosative Reactions in Cardiovascular Diseases. Front Physiol 2018; 9:672. [PMID: 29997515 PMCID: PMC6031070 DOI: 10.3389/fphys.2018.00672] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 05/15/2018] [Indexed: 11/16/2022] Open
Abstract
Metabolomics, also referred to as metabonomics, is one of the most recent innovative technologies in medicine. It offers a direct functional read-out of phenotypes by the detection, identification, and quantification of a large number of metabolites within a biological sample such as urine and blood. Metabolites (<1500 Da) represent the output of cellular metabolism, accounting for expression and activity of genes, transcripts, and proteins, and offering unique insights into small molecule regulation, which may uncover new biochemical patterns. Metabolomics research has considerable potential for translating the metabolic fingerprint into personalized therapeutic strategies. Within the field of interest, cardiovascular disease (CVD) is one of the most developed areas. However, CVD remains the leading cause of death worldwide with a marked increase in mortality rates over the past six decades. In this scenario, recent findings indicate the important role of redox and nitrosative (RN) reactions in CVD development and progression. RN reactions are generally involved in the homeostatic modulation of a wide number of cellular and organ functions. Conversely, the imbalance of these reactions may lead to a condition of allostasis that in turn can cause CVD. The aim of this review is to highlight how the use of metabolomics may be useful for the study of RN reactions related to CVD, providing a tool to understand the mechanisms underlying reactions that could lead to impaired ROS or RNS formation.
Collapse
Affiliation(s)
- Martino Deidda
- Department of Medical Sciences and Public Health, University of Cagliari, Sardinia, Italy
| | - Antonio Noto
- Department of Medical Sciences and Public Health, University of Cagliari, Sardinia, Italy
| | - Pier P Bassareo
- Department of Medical Sciences and Public Health, University of Cagliari, Sardinia, Italy
| | | | - Giuseppe Mercuro
- Department of Medical Sciences and Public Health, University of Cagliari, Sardinia, Italy
| |
Collapse
|
28
|
Santana MS, Nascimento KP, Lotufo PA, Benseãor IM, Meotti FC. Allantoin as an independent marker associated with carotid intima-media thickness in subclinical atherosclerosis. ACTA ACUST UNITED AC 2018; 51:e7543. [PMID: 29924138 PMCID: PMC6040864 DOI: 10.1590/1414-431x20187543] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 05/07/2018] [Indexed: 11/21/2022]
Abstract
Allantoin is the main product of uric acid oxidation and was found to be augmented in atherosclerotic plaque in human autopsy and in animal models of atherosclerosis. Uric acid is abundant in human plasma and is prone to oxidation in inflammatory conditions such as atherosclerosis. In this study, we found a significant increase in plasma uric acid (P=0.002) and allantoin (P=0.025) in participants of the Brazilian Longitudinal Study of Adult Health (ELSA-Brasil) that presented common carotid intima-media thickness (c-IMT) within the 75th percentile (c-IMT≥P75). Multiple linear regression showed an association of c-IMT with uric acid (β=0.0004, P=0.014) and allantoin (β=0.018, P=0.008). This association was independent of age, the traditional risk factor LDL/HDL ratio, and non-traditional risk factors: pulse pressure, neck circumference, and the inflammatory marker myeloperoxidase. The independent and strong association of allantoin with c-IMT shows that it might be a useful marker, along with other traditional risk factors, to evaluate an early stage of atherosclerosis.
Collapse
Affiliation(s)
- M S Santana
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, SP, Brasil
| | - K P Nascimento
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, SP, Brasil
| | - P A Lotufo
- Centro de Pesquisa Clínica e Epidemiológica, Hospital Universitário, Universidade de São Paulo, São Paulo, SP, Brasil
| | - I M Benseãor
- Centro de Pesquisa Clínica e Epidemiológica, Hospital Universitário, Universidade de São Paulo, São Paulo, SP, Brasil
| | - F C Meotti
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, SP, Brasil
| |
Collapse
|
29
|
Moss JWE, Williams JO, Ramji DP. Nutraceuticals as therapeutic agents for atherosclerosis. Biochim Biophys Acta Mol Basis Dis 2018; 1864:1562-1572. [PMID: 29454074 PMCID: PMC5906642 DOI: 10.1016/j.bbadis.2018.02.006] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 02/06/2018] [Accepted: 02/12/2018] [Indexed: 02/07/2023]
Abstract
Atherosclerosis, a chronic inflammatory disorder of medium and large arteries and an underlying cause of cardiovascular disease (CVD), is responsible for a third of all global deaths. Current treatments for CVD, such as optimized statin therapy, are associated with considerable residual risk and several side effects in some patients. The outcome of research on the identification of alternative pharmaceutical agents for the treatment of CVD has been relatively disappointing with many promising leads failing at the clinical level. Nutraceuticals, products from food sources with health benefits beyond their nutritional value, represent promising agents in the prevention of CVD or as an add-on therapy with current treatments. This review will highlight the potential of several nutraceuticals, including polyunsaturated fatty acids, flavonoids and other polyphenols, as anti-CVD therapies based on clinical and pre-clinical mechanism-based studies.
Collapse
Affiliation(s)
- Joe W E Moss
- Cardiff School of Biosciences, Cardiff University, Sir Martin Evans Building, Museum Avenue, Cardiff CF10 3AX, UK
| | - Jessica O Williams
- Cardiff School of Biosciences, Cardiff University, Sir Martin Evans Building, Museum Avenue, Cardiff CF10 3AX, UK
| | - Dipak P Ramji
- Cardiff School of Biosciences, Cardiff University, Sir Martin Evans Building, Museum Avenue, Cardiff CF10 3AX, UK.
| |
Collapse
|
30
|
Chen R, Wang J, Liao C, Zhang L, Guo Q, Wang X. Exploring the biomarkers and therapeutic mechanism of kidney-yang deficiency syndrome treated by You-gui pill using systems pharmacology and serum metabonomics. RSC Adv 2018; 8:1098-1115. [PMID: 35539000 PMCID: PMC9077015 DOI: 10.1039/c7ra12451a] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 12/11/2017] [Indexed: 01/07/2023] Open
Abstract
In this study, systems pharmacology was used to predict the molecular targets of You-gui pill (YGP) and explore the therapeutic mechanism of Kidney-Yang Deficiency Syndrome (KYDS) treated with YGP. On the basis of this, serum samples from control group, KYDS model group and YGP group rats were studied using 1H NMR to verify the results of systems pharmacology from the level of metabonomics. Simultaneously, 1H NMR spectra of serum samples were obtained and statistically assessed using pattern recognition analysis. Biochemical analyses of serums were performed via radioimmunoassays. Furthermore, histopathological studies were conducted on the pituitary, adrenal, and thyroid glands, and testicles of the control, KYDS and YGP rats. Using systems pharmacology to analyze the active components of YGP, 61 active compounds were finally found. These compounds were likely to have an effect on 3177 target proteins and involve 234 pathways. Using metabonomics to analyze serum from KYDS rats treated with YGP, 22 endogenous biomarkers were found. These biomarkers were mainly involved in 10 metabolic pathways. Combining systems pharmacology and metabonomics, we found that the regulation of KYDS was primarily associated with 19 active compounds of 5 Chinese herbal medicines in YGP. These active compounds mainly had an effect on 8 target proteins, including phosphoenolpyruvate carboxykinase, betaine-homocysteine s-methyltransferase 1, alcohol dehydrogenase 1C, etc. These target proteins were primarily involved in 6 overlapping pathways, namely aminoacyl-tRNA biosynthesis, glycolysis/gluconeogenesis, glycine, serine and threonine metabolism, valine, leucine and isoleucine biosynthesis, arginine and proline metabolism, and pyruvate metabolism. In addition, there were 4 non-overlapping pathways, respectively alanine, aspartate and glutamate metabolism, d-glutamine and d-glutamate metabolism, ubiquinone and other terpenoid-quinone biosynthesis, and galactose metabolism. In summary, the therapeutic effects of YGP on KYDS were mainly associated with neuroendocrine regulation, energy metabolism, amino acid metabolism, inflammatory responses, apoptosis, oxidative stress and intestinal flora metabolism. What's more, we also found that YGP possessed the potential to protect liver and kidney function. Our study demonstrated that systems pharmacology and metabonomics methods were novel strategies for the exploration of the mechanisms of KYDS and TCM formulas.
Collapse
Affiliation(s)
- Ruiqun Chen
- School of Basic Courses, Guangdong Pharmaceutical University Guangzhou 510006 P. R. China +86-20-39352186 +86-20-39352195
| | - Jia Wang
- School of Basic Courses, Guangdong Pharmaceutical University Guangzhou 510006 P. R. China +86-20-39352186 +86-20-39352195
| | - Chengbin Liao
- School of Basic Courses, Guangdong Pharmaceutical University Guangzhou 510006 P. R. China +86-20-39352186 +86-20-39352195
| | - Lei Zhang
- School of Basic Courses, Guangdong Pharmaceutical University Guangzhou 510006 P. R. China +86-20-39352186 +86-20-39352195
| | - Qian Guo
- School of Basic Courses, Guangdong Pharmaceutical University Guangzhou 510006 P. R. China +86-20-39352186 +86-20-39352195
| | - Xiufeng Wang
- School of Basic Courses, Guangdong Pharmaceutical University Guangzhou 510006 P. R. China +86-20-39352186 +86-20-39352195
| |
Collapse
|
31
|
Li B, Lu X, Wang J, He X, Gu Q, Wang L, Yang Y. The metabonomics study of P-selectin glycoprotein ligand-1 (PSGL-1) deficiency inhibiting the progression of atherosclerosis in LDLR -/- mice. Int J Biol Sci 2018; 14:36-46. [PMID: 29483823 PMCID: PMC5821047 DOI: 10.7150/ijbs.23082] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Accepted: 11/17/2017] [Indexed: 01/24/2023] Open
Abstract
Atherosclerosis (AS) is a multi-factorial chronic disease commonly associated with the mechanisms of metabolism disorder, endothelial dysfunction and chronic inflammation. AS an inflammatory molecule, p-selectin glycoprotein ligand-1 (PSGL-1) played an important role in the inflammatory process of atherogenesis involving the recruitment of leukocyte and transmitting signals to activate leukocyte during the adhesion process. So far, there has been little study regarding the effects of PSGL-1 on AS progression and the metabolic regulation. In this report, we studied the effect of PSGL-1 deficiency on the formation and progression of AS and the metabolic regulation by use of LDLR-/-, PSGL-1-/- transgenic mice based on metabonomics. It was found that the PSGL-1 deficiency reduced the atherosclerotic plaque area, inflammatory cells infiltration and fiber hyperplasia during the AS development. The serum metabonomics study showed that the LDLR-/- ,PSGL-1-/- mice had higher levels of HDL, valine, acetate, pyruvate, choline, PC, GPC and glycine, and lower levels of LDL+VLDL and lactate at the early stage of atherosclerosis, while lactate, citrate and glutamine showed statistical significance at the late stage of atherosclerosis. These results showed that the PSGL-1 deficiency inhibited the AS progression and regulated glucose metabolism, lipid metabolism, amino acid and phospholipid metabolism in LDLR-/- mice.
Collapse
Affiliation(s)
- Binglin Li
- Vascular Biology Research Institute, School of Basic Course, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Xin Lu
- Vascular Biology Research Institute, School of Basic Course, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Jia Wang
- Vascular Biology Research Institute, School of Basic Course, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Xiaodong He
- Vascular Biology Research Institute, School of Basic Course, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Quliang Gu
- Vascular Biology Research Institute, School of Basic Course, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Lijing Wang
- Vascular Biology Research Institute, School of Basic Course, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Yongxia Yang
- Vascular Biology Research Institute, School of Basic Course, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| |
Collapse
|
32
|
Myocardial metabolic alterations in mice with diet-induced atherosclerosis: linking sulfur amino acid and lipid metabolism. Sci Rep 2017; 7:13597. [PMID: 29051579 PMCID: PMC5648757 DOI: 10.1038/s41598-017-13991-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 10/04/2017] [Indexed: 02/08/2023] Open
Abstract
Atherosclerosis is a leading cause of cardiovascular disease (CVD), but the effect of diet on the atherosclerotic heart’s metabolism is unclear. We used an integrated metabolomics and lipidomics approach to evaluate metabolic perturbations in heart and serum from mice fed an atherogenic diet (AD) for 8, 16, and 25 weeks. Nuclear magnetic resonance (NMR)-based metabolomics revealed significant changes in sulfur amino acid (SAA) and lipid metabolism in heart from AD mice compared with heart from normal diet mice. Higher SAA levels in AD mice were quantitatively verified using liquid chromatography-mass spectrometry (LC/MS). Lipidomic profiling revealed that fatty acid and triglyceride (TG) levels in the AD group were altered depending on the degree of unsaturation. Additionally, levels of SCD1, SREBP-1, and PPARγ were reduced in AD mice after 25 weeks, while levels of reactive oxygen species were elevated. The results suggest that a long-term AD leads to SAA metabolism dysregulation and increased oxidative stress in the heart, causing SCD1 activity suppression and accumulation of toxic TGs with a low degree of unsaturation. These findings demonstrate that the SAA metabolic pathway is a promising therapeutic target for CVD treatment and that metabolomics can be used to investigate the metabolic signature of atherosclerosis.
Collapse
|
33
|
Yang Y, Zhang H, Yan B, Zhang T, Gao Y, Shi Y, Le G. Health Effects of Dietary Oxidized Tyrosine and Dityrosine Administration in Mice with Nutrimetabolomic Strategies. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2017; 65:6957-6971. [PMID: 28742334 DOI: 10.1021/acs.jafc.7b02003] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
This study aims to investigate the health effects of long-term dietary oxidized tyrosine (O-Tyr) and its main product (dityrosine) administration on mice metabolism. Mice received daily intragastric administration of either O-Tyr (320 μg/kg body weight), dityrosine (Dityr, 320 μg/kg body weight), or saline for consecutive 6 weeks. Urine and plasma samples were analyzed by NMR-based metabolomics strategies. Body weight, clinical chemistry, oxidative damage indexes, and histopathological data were obtained as complementary information. O-Tyr and Dityr exposure changed many systemic metabolic processes, including reduced choline bioavailability, led to fat accumulation in liver, induced hepatic injury, and renal dysfunction, resulted in changes in gut microbiota functions, elevated risk factor for cardiovascular disease, altered amino acid metabolism, induced oxidative stress responses, and inhibited energy metabolism. These findings implied that it is absolutely essential to reduce the generation of oxidation protein products in food system through improving modern food processing methods.
Collapse
Affiliation(s)
- Yuhui Yang
- The Laboratory of Food Nutrition and Functional Factors, School of Food Science and Technology, Jiangnan University , Wuxi, Jiangsu 214122, China
| | - Hui Zhang
- The Laboratory of Food Nutrition and Functional Factors, School of Food Science and Technology, Jiangnan University , Wuxi, Jiangsu 214122, China
| | - Biao Yan
- The Laboratory of Food Nutrition and Functional Factors, School of Food Science and Technology, Jiangnan University , Wuxi, Jiangsu 214122, China
| | - Tianyu Zhang
- The Laboratory of Food Nutrition and Functional Factors, School of Food Science and Technology, Jiangnan University , Wuxi, Jiangsu 214122, China
| | - Ying Gao
- The Laboratory of Food Nutrition and Functional Factors, School of Food Science and Technology, Jiangnan University , Wuxi, Jiangsu 214122, China
| | - Yonghui Shi
- The Laboratory of Food Nutrition and Functional Factors, School of Food Science and Technology, Jiangnan University , Wuxi, Jiangsu 214122, China
- The State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University , Wuxi, Jiangsu 214122, China
| | - Guowei Le
- The Laboratory of Food Nutrition and Functional Factors, School of Food Science and Technology, Jiangnan University , Wuxi, Jiangsu 214122, China
- The State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University , Wuxi, Jiangsu 214122, China
| |
Collapse
|
34
|
Wang J, Zhou L, Lei H, Hao F, Liu X, Wang Y, Tang H. Simultaneous Quantification of Amino Metabolites in Multiple Metabolic Pathways Using Ultra-High Performance Liquid Chromatography with Tandem-mass Spectrometry. Sci Rep 2017; 7:1423. [PMID: 28469184 PMCID: PMC5431165 DOI: 10.1038/s41598-017-01435-7] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 03/28/2017] [Indexed: 01/09/2023] Open
Abstract
Metabolites containing amino groups cover multiple pathways and play important roles in redox homeostasis and biosyntheses of proteins, nucleotides and neurotransmitters. Here, we report a new method for simultaneous quantification of 124 such metabolites. This is achieved by derivatization-assisted sensitivity enhancement with 5-aminoisoquinolyl-N-hydroxysuccinimidyl carbamate (5-AIQC) followed with comprehensive analysis using ultra-high performance liquid chromatography and electrospray ionization tandem mass spectrometry (UHPLC-MS/MS). In an one-pot manner, this quantification method enables simultaneous coverage of 20 important metabolic pathways including protein biosynthesis/degradation, biosyntheses of catecholamines, arginine and glutathione, metabolisms of homocysteine, taurine-hypotaurine etc. Compared with the reported ones, this method is capable of simultaneously quantifying thiols, disulfides and other oxidation-prone analytes in a single run and suitable for quantifying aromatic amino metabolites. This method is also much more sensitive for all tested metabolites with LODs well below 50 fmol (at sub-fmol for most tested analytes) and shows good precision for retention time and quantitation with inter-day and intra-day relative standard deviations (RSDs) below 15% and good recovery from renal cancer tissue, rat urine and plasma. The method was further applied to quantify the amino metabolites in silkworm hemolymph from multiple developmental stages showing its applicability in metabolomics and perhaps some clinical chemistry studies.
Collapse
Affiliation(s)
- Jin Wang
- School of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China.,State Key Laboratory of Genetic Engineering, Zhongshan Hospital and School of Life Sciences, Fudan University, Shanghai International Centre for Molecular Phenomics, Collaborative Innovation Center for Genetics and Development, Shanghai, 200438, China.,CAS Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Centre for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Lihong Zhou
- School of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China.,CAS Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Centre for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Hehua Lei
- CAS Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Centre for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Fuhua Hao
- CAS Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Centre for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Xin Liu
- School of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Yulan Wang
- CAS Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Centre for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan, 430071, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University, Hangzhou, 310058, China
| | - Huiru Tang
- State Key Laboratory of Genetic Engineering, Zhongshan Hospital and School of Life Sciences, Fudan University, Shanghai International Centre for Molecular Phenomics, Collaborative Innovation Center for Genetics and Development, Shanghai, 200438, China.
| |
Collapse
|
35
|
Cortese R, Gileles-Hillel A, Khalyfa A, Almendros I, Akbarpour M, Khalyfa AA, Qiao Z, Garcia T, Andrade J, Gozal D. Aorta macrophage inflammatory and epigenetic changes in a murine model of obstructive sleep apnea: Potential role of CD36. Sci Rep 2017; 7:43648. [PMID: 28240319 PMCID: PMC5327416 DOI: 10.1038/srep43648] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 01/25/2017] [Indexed: 12/15/2022] Open
Abstract
Obstructive sleep apnea (OSA) affects 8-10% of the population, is characterized by chronic intermittent hypoxia (CIH), and causally associates with cardiovascular morbidities. In CIH-exposed mice, closely mimicking the chronicity of human OSA, increased accumulation and proliferation of pro-inflammatory metabolic M1-like macrophages highly expressing CD36, emerged in aorta. Transcriptomic and MeDIP-seq approaches identified activation of pro-atherogenic pathways involving a complex interplay of histone modifications in functionally-relevant biological pathways, such as inflammation and oxidative stress in aorta macrophages. Discontinuation of CIH did not elicit significant improvements in aorta wall macrophage phenotype. However, CIH-induced aorta changes were absent in CD36 knockout mice, Our results provide mechanistic insights showing that CIH exposures during sleep in absence of concurrent pro-atherogenic settings (i.e., genetic propensity or dietary manipulation) lead to the recruitment of CD36(+)high macrophages to the aortic wall and trigger atherogenesis. Furthermore, long-term CIH-induced changes may not be reversible with usual OSA treatment.
Collapse
Affiliation(s)
- Rene Cortese
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL, USA
| | - Alex Gileles-Hillel
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL, USA
| | - Abdelnaby Khalyfa
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL, USA
| | - Isaac Almendros
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL, USA
| | - Mahzad Akbarpour
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL, USA
| | - Ahamed A Khalyfa
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL, USA
| | - Zhuanghong Qiao
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL, USA
| | - Tzintzuni Garcia
- Center for Research Informatics, The University of Chicago, Chicago, IL, USA
| | - Jorge Andrade
- Center for Research Informatics, The University of Chicago, Chicago, IL, USA
| | - David Gozal
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL, USA
| |
Collapse
|
36
|
Wan C, Zhan Y, Xue R, Wu Y, Li X, Pei F. Gd-DTPA-induced dynamic metabonomic changes in rat biofluids. Magn Reson Imaging 2017; 44:15-25. [PMID: 28095303 DOI: 10.1016/j.mri.2017.01.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 01/09/2017] [Accepted: 01/09/2017] [Indexed: 01/22/2023]
Abstract
OBJECTIVES The purposes of this study were (1) to detect the dynamic metabonomic changes induced by gadopentetate dimeglumine (Gd-DTPA) and (2) to investigate the potential metabolic disturbances associated with the pathogenesis of nephrogenic systemic fibrosis (NSF) at the early stage. METHODS A nuclear magnetic resonance (NMR)-based metabolomics approach was used to investigate the urinary and serum metabolic changes induced by a single tail vein injection of Gd-DTPA (dosed at 2 and 5mmol/kg body weight) in rats. Urine and serum samples were collected on days 1, 2 and 7 after dosing. RESULTS Metabolic responses of rats to Gd-DTPA administration were systematic involving changes in lipid metabolism, glucose metabolism, TCA cycle, amino acid metabolism and gut microbiota functions. Urinary and serum metabonomic recovery could be observed in both the 2 and 5mmol/kg body weight group, but the metabolic effects of high-dosed (5mmol/kg body weight) Gd-DTPA lasted longer. It is worth noting that hyperlipidemia was observed after Gd-DTPA injection, and nicotinate might play a role in the subsequent self-recovery of lipid metabolism. The disturbance of tyrosine, glutamate and gut microbiota metabolism might associate with the progression of NSF. CONCLUSION These findings offered essential information about the metabolic changes induced by Gd-DTPA, and could be potentially important for investigating the pathogenesis of NSF at the early stage. Moreover, the recovery of rats administrated with Gd-DTPA may have implications in the treatment of early stage NSF.
Collapse
Affiliation(s)
- Chuanling Wan
- Changchun Institute of Applied Chemistry Chinese Academy of Sciences, No. 5625, Renmin Street, Changchun 130022, China; University of Chinese Academy of Sciences, No. 19, Yuquan Road 19, Beijing 100049, China
| | - Youyang Zhan
- Changchun Institute of Applied Chemistry Chinese Academy of Sciences, No. 5625, Renmin Street, Changchun 130022, China; University of Chinese Academy of Sciences, No. 19, Yuquan Road 19, Beijing 100049, China
| | - Rong Xue
- Changchun Institute of Applied Chemistry Chinese Academy of Sciences, No. 5625, Renmin Street, Changchun 130022, China
| | - Yijie Wu
- Changchun Institute of Applied Chemistry Chinese Academy of Sciences, No. 5625, Renmin Street, Changchun 130022, China
| | - Xiaojing Li
- Changchun Institute of Applied Chemistry Chinese Academy of Sciences, No. 5625, Renmin Street, Changchun 130022, China.
| | - Fengkui Pei
- Changchun Institute of Applied Chemistry Chinese Academy of Sciences, No. 5625, Renmin Street, Changchun 130022, China
| |
Collapse
|
37
|
Lin L, Zhang J. Role of intestinal microbiota and metabolites on gut homeostasis and human diseases. BMC Immunol 2017; 18:2. [PMID: 28061847 PMCID: PMC5219689 DOI: 10.1186/s12865-016-0187-3] [Citation(s) in RCA: 443] [Impact Index Per Article: 55.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 12/20/2016] [Indexed: 12/12/2022] Open
Abstract
Background A vast diversity of microbes colonizes in the human gastrointestinal tract, referred to intestinal microbiota. Microbiota and products thereof are indispensable for shaping the development and function of host innate immune system, thereby exerting multifaceted impacts in gut health. Methods This paper reviews the effects on immunity of gut microbe-derived nucleic acids, and gut microbial metabolites, as well as the involvement of commensals in the gut homeostasis. We focus on the recent findings with an intention to illuminate the mechanisms by which the microbiota and products thereof are interacting with host immunity, as well as to scrutinize imbalanced gut microbiota (dysbiosis) which lead to autoimmune disorders including inflammatory bowel disease (IBD), Type 1 diabetes (T1D) and systemic immune syndromes such as rheumatoid arthritis (RA). Results In addition to their well-recognized benefits in the gut such as occupation of ecological niches and competition with pathogens, commensal bacteria have been shown to strengthen the gut barrier and to exert immunomodulatory actions within the gut and beyond. It has been realized that impaired intestinal microbiota not only contribute to gut diseases but also are inextricably linked to metabolic disorders and even brain dysfunction. Conclusions A better understanding of the mutual interactions of the microbiota and host immune system, would shed light on our endeavors of disease prevention and broaden the path to our discovery of immune intervention targets for disease treatment.
Collapse
Affiliation(s)
- Lan Lin
- Department of Bioengineering, Medical School, Southeast University, Nanjing, 210009, People's Republic of China.
| | - Jianqiong Zhang
- Key Laboratory of Developmental Genes and Human Disease, Ministry of Education, Department of Microbiology and Immunology, Medical School, Southeast University, Nanjing, 210009, People's Republic of China.
| |
Collapse
|
38
|
Yang Y, Zhang J, Liu Y, Li B, Li J, Zheng L, Wang L. Metabonomic analysis of metastatic lung tissue in breast cancer mice by an integrated NMR-based metabonomics approach. RSC Adv 2017. [DOI: 10.1039/c7ra02069d] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
This study identified the common potential biomarkers for early lung metastasis of breast cancer in two models.
Collapse
Affiliation(s)
- Yongxia Yang
- School of Basic Course
- Guangdong Pharmaceutical University
- Guangzhou
- PR China
- Vascular Biology Research Institute
| | - Jingli Zhang
- School of Basic Course
- Guangdong Pharmaceutical University
- Guangzhou
- PR China
- Vascular Biology Research Institute
| | - Ying Liu
- Vascular Biology Research Institute
- Guangdong Pharmaceutical University
- Guangzhou
- PR China
| | - Binglin Li
- School of Basic Course
- Guangdong Pharmaceutical University
- Guangzhou
- PR China
- Vascular Biology Research Institute
| | - Jiangchao Li
- Vascular Biology Research Institute
- Guangdong Pharmaceutical University
- Guangzhou
- PR China
| | - Lingyun Zheng
- School of Basic Course
- Guangdong Pharmaceutical University
- Guangzhou
- PR China
| | - Lijing Wang
- Vascular Biology Research Institute
- Guangdong Pharmaceutical University
- Guangzhou
- PR China
| |
Collapse
|
39
|
Yang Y, Yan B, Cheng X, Ding Y, Tian X, Shi Y, Le G. Metabolomic studies on the systemic responses of mice with oxidative stress induced by short-term oxidized tyrosine administration. RSC Adv 2017. [DOI: 10.1039/c7ra02665j] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Oxidized tyrosine (O-Tyr) has attracted more interest in recent years because many researchers have discovered that it and its product (dityrosine) are associated with pathological conditions, especially various age-related disorders in biological systems.
Collapse
Affiliation(s)
- Yuhui Yang
- The Laboratory of Food Nutrition and Functional Factors
- School of Food Science and Technology
- Jiangnan University
- Wuxi 214122
- China
| | - Biao Yan
- The Laboratory of Food Nutrition and Functional Factors
- School of Food Science and Technology
- Jiangnan University
- Wuxi 214122
- China
| | - Xiangrong Cheng
- The Laboratory of Food Nutrition and Functional Factors
- School of Food Science and Technology
- Jiangnan University
- Wuxi 214122
- China
| | - Yinyi Ding
- The Laboratory of Food Nutrition and Functional Factors
- School of Food Science and Technology
- Jiangnan University
- Wuxi 214122
- China
| | - Xu Tian
- The Laboratory of Food Nutrition and Functional Factors
- School of Food Science and Technology
- Jiangnan University
- Wuxi 214122
- China
| | - Yonghui Shi
- The Laboratory of Food Nutrition and Functional Factors
- School of Food Science and Technology
- Jiangnan University
- Wuxi 214122
- China
| | - Guowei Le
- The Laboratory of Food Nutrition and Functional Factors
- School of Food Science and Technology
- Jiangnan University
- Wuxi 214122
- China
| |
Collapse
|
40
|
Abstract
Infections have been linked to the development of cardiovascular disease and atherosclerosis. Findings from the past decade have identified microbial ecosystems residing in different habitats of the human body that contribute to metabolic and cardiovascular-related disorders. In this Review, we describe three pathways by which microbiota might affect atherogenesis. First, local or distant infections might cause a harmful inflammatory response that aggravates plaque development or triggers plaque rupture. Second, metabolism of cholesterol and lipids by gut microbiota can affect the development of atherosclerotic plaques. Third, diet and specific components that are metabolized by gut microbiota can have various effects on atherosclerosis; for example, dietary fibre is beneficial, whereas the bacterial metabolite trimethylamine-N-oxide is considered harmful. Although specific bacterial taxa have been associated with atherosclerosis, which is supported by increasing mechanistic evidence, several questions remain to be answered to understand fully how the microbiota contributes to atherosclerosis and cardiovascular disease. Such knowledge might pave the way for novel diagnostics and therapeutics based on microbiota.
Collapse
Affiliation(s)
- Annika Lindskog Jonsson
- Wallenberg Laboratory for Cardiovascular and Metabolic Research, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Bruna Stråket 16, 41345 Gothenburg, Sweden
| | - Fredrik Bäckhed
- Wallenberg Laboratory for Cardiovascular and Metabolic Research, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Bruna Stråket 16, 41345 Gothenburg, Sweden.,Novo Nordisk Foundation Center for Basic Metabolic Research, Section for Metabolic Receptology and Enteroendocrinology, Faculty of Health Sciences, University of Copenhagen, Copenhagen, DK-2200, Denmark
| |
Collapse
|
41
|
Bao MH, Luo HQ, Chen LH, Tang L, Ma KF, Xiang J, Dong LP, Zeng J, Li GY, Li JM. Impact of high fat diet on long non-coding RNAs and messenger RNAs expression in the aortas of ApoE(-/-) mice. Sci Rep 2016; 6:34161. [PMID: 27698357 PMCID: PMC5048419 DOI: 10.1038/srep34161] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 09/07/2016] [Indexed: 12/13/2022] Open
Abstract
Atherosclerosis is a chronic multifactorial inflammatory disease with high prevalence worldwide, and has become the leading cause of death. The present study was designed to investigate the impact of high-fat diet on ApoE(−/−) mice exhibiting atherosclerosis by detecting the genome-wide expression profile of lncRNAs and mRNAs. A total of 354 differentially expressed lncRNAs were identified (≥2.0 folds). Simultaneously, 357 differentially expressed mRNAs from the same chip were found. The expression differences of lncRNAs and mRNAs were consistent in both qPCR and microarray detection. Annotation results of the mRNAs which correlated with lncRNAs showed that the commonly related pathways were metabolism and inflammation. Hypergeometric distribution analysis indicated that the differentially expressed lncRNAs had been mostly regulated by transcription factors (TFs) such as Myod1, Rxra, Pparg, Tcf3, etc. Additional lncRNA-target-TFs network analysis was conducted for the top 20 differentially expressed lncRNAs. The results indicated Hnf4a, Ppara, Vdr, and Runx3 as the TFs most likely to regulate the production of these lncRNAs, and might play roles in inflammatory and metabolic processes in atherosclerosis. In a nutshell, the present study identified a panel of dysregulated lncRNAs and mRNAs that may be potential biomarkers or drug targets relevant to the high-fat diet related atherogenesis.
Collapse
Affiliation(s)
- Mei-Hua Bao
- Department of Anatomy, Histology and Embryology, Institute of of Neuroscience, Changsha Medical University, Changsha, 410219, China
| | - Huai-Qing Luo
- Department of Anatomy, Histology and Embryology, Institute of of Neuroscience, Changsha Medical University, Changsha, 410219, China
| | - Li-Hua Chen
- The Third Xiangya Hospital of Central South University, Changsha 410013, Hunan, PR China
| | - Liang Tang
- Department of Anatomy, Histology and Embryology, Institute of of Neuroscience, Changsha Medical University, Changsha, 410219, China
| | - Kui-Fen Ma
- The First Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang, PR China
| | - Ju Xiang
- Department of Anatomy, Histology and Embryology, Institute of of Neuroscience, Changsha Medical University, Changsha, 410219, China
| | - Li-Ping Dong
- Department of Anatomy, Histology and Embryology, Institute of of Neuroscience, Changsha Medical University, Changsha, 410219, China
| | - Jie Zeng
- Department of Anatomy, Histology and Embryology, Institute of of Neuroscience, Changsha Medical University, Changsha, 410219, China
| | - Guang-Yi Li
- Department of Anatomy, Histology and Embryology, Institute of of Neuroscience, Changsha Medical University, Changsha, 410219, China
| | - Jian-Ming Li
- Department of Anatomy, Histology and Embryology, Institute of of Neuroscience, Changsha Medical University, Changsha, 410219, China.,Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China.,Department of Anatomy, Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, China
| |
Collapse
|
42
|
Li H, Stokes W, Chater E, Roy R, de Bruin E, Hu Y, Liu Z, Smit EF, Heynen GJJE, Downward J, Seckl MJ, Wang Y, Tang H, Pardo OE. Decreased glutathione biosynthesis contributes to EGFR T790M-driven erlotinib resistance in non-small cell lung cancer. Cell Discov 2016; 2:16031. [PMID: 27721983 PMCID: PMC5037574 DOI: 10.1038/celldisc.2016.31] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Accepted: 07/14/2016] [Indexed: 12/11/2022] Open
Abstract
Epidermal growth factor receptor (EGFR) inhibitors such as erlotinib are novel effective agents in the treatment of EGFR-driven lung cancer, but their clinical impact is often impaired by acquired drug resistance through the secondary T790M EGFR mutation. To overcome this problem, we analysed the metabonomic differences between two independent pairs of erlotinib-sensitive/resistant cells and discovered that glutathione (GSH) levels were significantly reduced in T790M EGFR cells. We also found that increasing GSH levels in erlotinib-resistant cells re-sensitised them, whereas reducing GSH levels in erlotinib-sensitive cells made them resistant. Decreased transcription of the GSH-synthesising enzymes (GCLC and GSS) due to the inhibition of NRF2 was responsible for low GSH levels in resistant cells that was directly linked to the T790M mutation. T790M EGFR clinical samples also showed decreased expression of these key enzymes; increasing intra-tumoural GSH levels with a small-molecule GST inhibitor re-sensitised resistant tumours to erlotinib in mice. Thus, we identified a new resistance pathway controlled by EGFR T790M and a therapeutic strategy to tackle this problem in the clinic.
Collapse
Affiliation(s)
- Hongde Li
- State Key Laboratory of Genetic Engineering, Ministry of Education Key Laboratory of Contemporary Anthropology, Collaborative Innovation Centre for Genetics and Development, Shanghai International Centre for Molecular Phenomics, Metabonomics and Systems Biology Laboratory, Department of Biochemistry, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, China
- Key Laboratory of Magnetic Resonance in Biological Systems, National Centre for Magnetic Resonance in Wuhan, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan, China
| | - William Stokes
- Division of Cancer, Department of Surgery and Cancer, Imperial College, Hammersmith Hospital, London, UK
| | - Emily Chater
- Division of Cancer, Department of Surgery and Cancer, Imperial College, Hammersmith Hospital, London, UK
| | - Rajat Roy
- Division of Cancer, Department of Surgery and Cancer, Imperial College, Hammersmith Hospital, London, UK
| | - Elza de Bruin
- Signal Transduction Laboratory, CRUK London Research Institute, London, UK
- Personalised Healthcare & Biomarkers, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Cambridge, UK
| | - Yili Hu
- Key Laboratory of Magnetic Resonance in Biological Systems, National Centre for Magnetic Resonance in Wuhan, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan, China
| | - Zhigang Liu
- Key Laboratory of Magnetic Resonance in Biological Systems, National Centre for Magnetic Resonance in Wuhan, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan, China
| | - Egbert F Smit
- Department of Pulmonary Diseases, VU University Medical Centre, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Guus JJE Heynen
- Section of Molecular Carcinogenesis, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Julian Downward
- Signal Transduction Laboratory, CRUK London Research Institute, London, UK
| | - Michael J Seckl
- Division of Cancer, Department of Surgery and Cancer, Imperial College, Hammersmith Hospital, London, UK
| | - Yulan Wang
- Key Laboratory of Magnetic Resonance in Biological Systems, National Centre for Magnetic Resonance in Wuhan, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan, China
- Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Huiru Tang
- State Key Laboratory of Genetic Engineering, Ministry of Education Key Laboratory of Contemporary Anthropology, Collaborative Innovation Centre for Genetics and Development, Shanghai International Centre for Molecular Phenomics, Metabonomics and Systems Biology Laboratory, Department of Biochemistry, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, China
- Key Laboratory of Magnetic Resonance in Biological Systems, National Centre for Magnetic Resonance in Wuhan, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan, China
| | - Olivier E Pardo
- Division of Cancer, Department of Surgery and Cancer, Imperial College, Hammersmith Hospital, London, UK
| |
Collapse
|
43
|
Guo W, Jiang C, Yang L, Li T, Liu X, Jin M, Qu K, Chen H, Jin X, Liu H, Zhu H, Wang Y. Quantitative Metabolomic Profiling of Plasma, Urine, and Liver Extracts by 1H NMR Spectroscopy Characterizes Different Stages of Atherosclerosis in Hamsters. J Proteome Res 2016; 15:3500-3510. [PMID: 27570155 DOI: 10.1021/acs.jproteome.6b00179] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Atherosclerosis (AS) is a progressive disease that contributes to cardiovascular disease and shows a complex etiology, including genetic and environmental factors. To understand systemic metabolic changes and to identify potential biomarkers correlated with the occurrence and perpetuation of diet-induced AS, we applied 1H NMR-based metabolomics to detect the time-related metabolic profiles of plasma, urine, and liver extracts from male hamsters fed a high fat and high cholesterol (HFHC) diet. Conventional biochemical assays and histopathological examinations as well as protein expression analyses were performed to provide complementary information. We found that diet treatment caused obvious aortic lesions, lipid accumulation, and inflammatory infiltration in hamsters. Downregulation of proteins related to cholesterol metabolism, including hepatic SREBP2, LDL-R, CYP7A1, SR-BI, HMGCR, LCAT, and SOAT1 was detected, which elucidated the perturbation of cholesterol homeostasis during the HFHC diet challenge. Using "targeted analysis", we quantified 40 plasma, 80 urine, and 60 liver hydrophilic extract metabolites. Multivariate analyses of the identified metabolites elucidated sophisticated metabolic disturbances in multiple matrices, including energy homeostasis, intestinal microbiota functions, inflammation, and oxidative stress coupled with the metabolisms of cholesterol, fatty acids, saccharides, choline, amino acids, and nucleotides. For the first time, our results demonstrate a time-dependent metabolic progression of multiple biological matrices in hamsters from physiological status to early AS and further to late-stage AS, demonstrating that 1H NMR-based metabolomics is a reliable tool for early diagnosis and monitoring of the process of AS.
Collapse
Affiliation(s)
- Wei Guo
- State Key Laboratory for Bioactive Substances and Functions of Natural Medicines and ‡Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College , No.1 Xiannongtan Street, Beijing 100050, P. R. China
| | - Chunying Jiang
- State Key Laboratory for Bioactive Substances and Functions of Natural Medicines and ‡Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College , No.1 Xiannongtan Street, Beijing 100050, P. R. China
| | - Liu Yang
- State Key Laboratory for Bioactive Substances and Functions of Natural Medicines and ‡Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College , No.1 Xiannongtan Street, Beijing 100050, P. R. China
| | - Tianqi Li
- State Key Laboratory for Bioactive Substances and Functions of Natural Medicines and ‡Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College , No.1 Xiannongtan Street, Beijing 100050, P. R. China
| | - Xia Liu
- State Key Laboratory for Bioactive Substances and Functions of Natural Medicines and ‡Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College , No.1 Xiannongtan Street, Beijing 100050, P. R. China
| | - Mengxia Jin
- State Key Laboratory for Bioactive Substances and Functions of Natural Medicines and ‡Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College , No.1 Xiannongtan Street, Beijing 100050, P. R. China
| | - Kai Qu
- State Key Laboratory for Bioactive Substances and Functions of Natural Medicines and ‡Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College , No.1 Xiannongtan Street, Beijing 100050, P. R. China
| | - Huili Chen
- State Key Laboratory for Bioactive Substances and Functions of Natural Medicines and ‡Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College , No.1 Xiannongtan Street, Beijing 100050, P. R. China
| | - Xiangju Jin
- State Key Laboratory for Bioactive Substances and Functions of Natural Medicines and ‡Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College , No.1 Xiannongtan Street, Beijing 100050, P. R. China
| | - Hongyue Liu
- State Key Laboratory for Bioactive Substances and Functions of Natural Medicines and ‡Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College , No.1 Xiannongtan Street, Beijing 100050, P. R. China
| | - Haibo Zhu
- State Key Laboratory for Bioactive Substances and Functions of Natural Medicines and ‡Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College , No.1 Xiannongtan Street, Beijing 100050, P. R. China
| | - Yinghong Wang
- State Key Laboratory for Bioactive Substances and Functions of Natural Medicines and ‡Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College , No.1 Xiannongtan Street, Beijing 100050, P. R. China
| |
Collapse
|
44
|
Pelantová H, Bugáňová M, Holubová M, Šedivá B, Zemenová J, Sýkora D, Kaválková P, Haluzík M, Železná B, Maletínská L, Kuneš J, Kuzma M. Urinary metabolomic profiling in mice with diet-induced obesity and type 2 diabetes mellitus after treatment with metformin, vildagliptin and their combination. Mol Cell Endocrinol 2016; 431:88-100. [PMID: 27164444 DOI: 10.1016/j.mce.2016.05.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2016] [Revised: 04/15/2016] [Accepted: 05/05/2016] [Indexed: 01/06/2023]
Abstract
Metformin, vildagliptin and their combination are widely used for the treatment of diabetes, but little is known about the metabolic responses to these treatments. In the present study, NMR-based metabolomics was applied to detect changes in the urinary metabolomic profile of a mouse model of diet-induced obesity in response to these treatments. Additionally, standard biochemical parameters and the expression of enzymes involved in glucose and fat metabolism were monitored. Significant correlations were observed between several metabolites (e.g., N-carbamoyl-β-alanine, N1-methyl-4-pyridone-3-carboxamide, N1-methyl-2-pyridone-5-carboxamide, glucose, 3-indoxyl sulfate, dimethylglycine and several acylglycines) and the area under the curve of glucose concentrations during the oral glucose tolerance test. The present study is the first to present N-carbamoyl-β-alanine as a potential marker of type 2 diabetes mellitus and consequently to demonstrate the efficacies of the applied antidiabetic interventions. Moreover, the elevated acetate level observed after vildagliptin administration might reflect increased fatty acid oxidation.
Collapse
Affiliation(s)
- Helena Pelantová
- Institute of Microbiology, Academy of Sciences of the Czech Republic, Vídeňská 1083, 142 20, Prague 4, Czech Republic; Department of Analytical Chemistry, Faculty of Science, Palacký University, 17 listopadu 1192/12, 771 46, Olomouc, Czech Republic
| | - Martina Bugáňová
- Institute of Microbiology, Academy of Sciences of the Czech Republic, Vídeňská 1083, 142 20, Prague 4, Czech Republic; Faculty of Chemical Technology, University of Chemistry and Technology Prague, Technická 5, 166 28, Prague 6, Czech Republic
| | - Martina Holubová
- Institute of Organic Chemistry and Biochemistry, Academy of Sciences of the Czech Republic, Flemingovo nám. 2, 166 10, Prague 6, Czech Republic
| | - Blanka Šedivá
- Faculty of Applied Sciences, University of West Bohemia, Univerzitní 8, 306 14, Plzeň, Czech Republic
| | - Jana Zemenová
- Institute of Organic Chemistry and Biochemistry, Academy of Sciences of the Czech Republic, Flemingovo nám. 2, 166 10, Prague 6, Czech Republic; Faculty of Chemical Engineering, University of Chemistry and Technology Prague, Technická 3, 166 28, Prague 6, Czech Republic
| | - David Sýkora
- Faculty of Chemical Engineering, University of Chemistry and Technology Prague, Technická 3, 166 28, Prague 6, Czech Republic
| | - Petra Kaválková
- Institute of Medical Biochemistry and Laboratory Diagnostics, 1st Faculty of Medicine, Charles University and General Faculty Hospital in Prague, U nemocnice 1, 128 08, Prague 2, Czech Republic
| | - Martin Haluzík
- Institute of Medical Biochemistry and Laboratory Diagnostics, 1st Faculty of Medicine, Charles University and General Faculty Hospital in Prague, U nemocnice 1, 128 08, Prague 2, Czech Republic; Institute of Endocrinology, Národní 8, 116 94, Prague 1, Czech Republic
| | - Blanka Železná
- Institute of Organic Chemistry and Biochemistry, Academy of Sciences of the Czech Republic, Flemingovo nám. 2, 166 10, Prague 6, Czech Republic
| | - Lenka Maletínská
- Institute of Organic Chemistry and Biochemistry, Academy of Sciences of the Czech Republic, Flemingovo nám. 2, 166 10, Prague 6, Czech Republic
| | - Jaroslav Kuneš
- Institute of Organic Chemistry and Biochemistry, Academy of Sciences of the Czech Republic, Flemingovo nám. 2, 166 10, Prague 6, Czech Republic; Institute of Physiology, Academy of Sciences of the Czech Republic, Vídeňská 1083, 142 20, Prague 4, Czech Republic
| | - Marek Kuzma
- Institute of Microbiology, Academy of Sciences of the Czech Republic, Vídeňská 1083, 142 20, Prague 4, Czech Republic.
| |
Collapse
|
45
|
Tumor growth affects the metabonomic phenotypes of multiple mouse non-involved organs in an A549 lung cancer xenograft model. Sci Rep 2016; 6:28057. [PMID: 27329570 PMCID: PMC4916411 DOI: 10.1038/srep28057] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2016] [Accepted: 05/31/2016] [Indexed: 02/05/2023] Open
Abstract
The effects of tumorigenesis and tumor growth on the non-involved organs remain poorly understood although many research efforts have already been made for understanding the metabolic phenotypes of various tumors. To better the situation, we systematically analyzed the metabolic phenotypes of multiple non-involved mouse organ tissues (heart, liver, spleen, lung and kidney) in an A549 lung cancer xenograft model at two different tumor-growth stages using the NMR-based metabonomics approaches. We found that tumor growth caused significant metabonomic changes in multiple non-involved organ tissues involving numerous metabolic pathways, including glycolysis, TCA cycle and metabolisms of amino acids, fatty acids, choline and nucleic acids. Amongst these, the common effects are enhanced glycolysis and nucleoside/nucleotide metabolisms. These findings provided essential biochemistry information about the effects of tumor growth on the non-involved organs.
Collapse
|
46
|
Wu J, Yang L, Li S, Huang P, Liu Y, Wang Y, Tang H. Metabolomics Insights into the Modulatory Effects of Long-Term Low Calorie Intake in Mice. J Proteome Res 2016; 15:2299-308. [DOI: 10.1021/acs.jproteome.6b00336] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Junfang Wu
- Key
Laboratory of Magnetic Resonance in Biological Systems, State Key
Laboratory of Magnetic Resonance and Atomic and Molecular Physics,
Wuhan Centre for Magnetic Resonance, Wuhan Institute of Physics and
Mathematics, Chinese Academy of Sciences, Wuhan 430071, P. R. China
| | - Liu Yang
- Key
Laboratory of Nutrition and Metabolism, Institute for Nutritional
Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, P. R. China
| | - Shoufeng Li
- Key
Laboratory of Nutrition and Metabolism, Institute for Nutritional
Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, P. R. China
| | - Ping Huang
- Key
Laboratory of Nutrition and Metabolism, Institute for Nutritional
Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, P. R. China
| | - Yong Liu
- Key
Laboratory of Nutrition and Metabolism, Institute for Nutritional
Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, P. R. China
| | - Yulan Wang
- Key
Laboratory of Magnetic Resonance in Biological Systems, State Key
Laboratory of Magnetic Resonance and Atomic and Molecular Physics,
Wuhan Centre for Magnetic Resonance, Wuhan Institute of Physics and
Mathematics, Chinese Academy of Sciences, Wuhan 430071, P. R. China
- Collaborative
Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University, Hangzhou 310058, P. R. China
| | - Huiru Tang
- Key
Laboratory of Magnetic Resonance in Biological Systems, State Key
Laboratory of Magnetic Resonance and Atomic and Molecular Physics,
Wuhan Centre for Magnetic Resonance, Wuhan Institute of Physics and
Mathematics, Chinese Academy of Sciences, Wuhan 430071, P. R. China
- State Key
Laboratory of Genetic Engineering, Collaborative Innovation Center
for Genetics and Development, Metabolomics and Systems Biology Laboratory,
School of Life Sciences, Fudan University, Shanghai 200433, P. R. China
| |
Collapse
|
47
|
Siderophore biosynthesis coordinately modulated the virulence-associated interactive metabolome of uropathogenic Escherichia coli and human urine. Sci Rep 2016; 6:24099. [PMID: 27076285 PMCID: PMC4831015 DOI: 10.1038/srep24099] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Accepted: 03/22/2016] [Indexed: 02/06/2023] Open
Abstract
Uropathogenic Escherichia coli (UPEC) growth in women’s bladders during urinary tract infection (UTI) incurs substantial chemical exchange, termed the “interactive metabolome”, which primarily accounts for the metabolic costs (utilized metabolome) and metabolic donations (excreted metabolome) between UPEC and human urine. Here, we attempted to identify the individualized interactive metabolome between UPEC and human urine. We were able to distinguish UPEC from non-UPEC by employing a combination of metabolomics and genetics. Our results revealed that the interactive metabolome between UPEC and human urine was markedly different from that between non-UPEC and human urine, and that UPEC triggered much stronger perturbations in the interactive metabolome in human urine. Furthermore, siderophore biosynthesis coordinately modulated the individualized interactive metabolome, which we found to be a critical component of UPEC virulence. The individualized virulence-associated interactive metabolome contained 31 different metabolites and 17 central metabolic pathways that were annotated to host these different metabolites, including energetic metabolism, amino acid metabolism, and gut microbe metabolism. Changes in the activities of these pathways mechanistically pinpointed the virulent capability of siderophore biosynthesis. Together, our findings provide novel insights into UPEC virulence, and we propose that siderophores are potential targets for further discovery of drugs to treat UPEC-induced UTI.
Collapse
|
48
|
Su Q, Guan T, He Y, Lv H. Siderophore Biosynthesis Governs the Virulence of Uropathogenic Escherichia coli by Coordinately Modulating the Differential Metabolism. J Proteome Res 2016; 15:1323-32. [DOI: 10.1021/acs.jproteome.6b00061] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Qiao Su
- The Laboratory
for Functional
Omics and Innovative Chinese Medicine, Innovative Drug Research Center, Chongqing University, Chongqing 401331, P.R. China
| | - Tianbing Guan
- The Laboratory
for Functional
Omics and Innovative Chinese Medicine, Innovative Drug Research Center, Chongqing University, Chongqing 401331, P.R. China
| | - Yan He
- The Laboratory
for Functional
Omics and Innovative Chinese Medicine, Innovative Drug Research Center, Chongqing University, Chongqing 401331, P.R. China
| | - Haitao Lv
- The Laboratory
for Functional
Omics and Innovative Chinese Medicine, Innovative Drug Research Center, Chongqing University, Chongqing 401331, P.R. China
| |
Collapse
|
49
|
Correlations of Fecal Metabonomic and Microbiomic Changes Induced by High-fat Diet in the Pre-Obesity State. Sci Rep 2016; 6:21618. [PMID: 26916743 PMCID: PMC4768318 DOI: 10.1038/srep21618] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 01/26/2016] [Indexed: 02/07/2023] Open
Abstract
Obesity resulting from interactions of genetic and environmental factors becomes a serious public health problem worldwide with alterations of the metabolic phenotypes in multiple biological matrices involving multiple metabolic pathways. To understand the contributions of gut microbiota to obesity development, we analyzed dynamic alterations in fecal metabonomic phenotype using NMR and fecal microorganism composition in rats using pyrosequencing technology during the high-fat diet (HFD) feeding for 81 days (pre-obesity state). Integrated analysis of these two phenotypic datasets was further conducted to establish correlations between the altered rat fecal metabonome and gut microbiome. We found that one-week HFD feeding already caused significant changes in rat fecal metabonome and such changes sustained throughout 81-days feeding with the host and gut microbiota co-metabolites clearly featured. We also found that HFD caused outstanding decreases in most fecal metabolites implying enhancement of gut absorptions. We further established comprehensive correlations between the HFD-induced changes in fecal metabonome and fecal microbial composition indicating contributions of gut microbiota in pathogenesis and progression of the HFD-induced obesity. These findings provided essential information about the functions of gut microbiota in pathogenesis of metabolic disorders which could be potentially important for developing obesity prevention and treatment therapies.
Collapse
|
50
|
Liu C, Ding F, Hao F, Yu M, Lei H, Wu X, Zhao Z, Guo H, Yin J, Wang Y, Tang H. Reprogramming of Seed Metabolism Facilitates Pre-harvest Sprouting Resistance of Wheat. Sci Rep 2016; 6:20593. [PMID: 26860057 PMCID: PMC4748292 DOI: 10.1038/srep20593] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 01/07/2016] [Indexed: 12/14/2022] Open
Abstract
Pre-harvest sprouting (PHS) is a worldwide problem for wheat production and transgene antisense-thioredoxin-s (anti-trx-s) facilitates outstanding resistance. To understand the molecular details of PHS resistance, we analyzed the metabonomes of the transgenic and wild-type (control) wheat seeds at various stages using NMR and GC-FID/MS. 60 metabolites were dominant in these seeds including sugars, organic acids, amino acids, choline metabolites and fatty acids. At day-20 post-anthesis, only malate level in transgenic wheat differed significantly from that in controls whereas at day-30 post-anthesis, levels of amino acids and sucrose were significantly different between these two groups. For mature seeds, most metabolites in glycolysis, TCA cycle, choline metabolism, biosynthesis of proteins, nucleotides and fatty acids had significantly lower levels in transgenic seeds than in controls. After 30-days post-harvest ripening, most metabolites in transgenic seeds had higher levels than in controls including amino acids, sugars, organic acids, fatty acids, choline metabolites and NAD+. These indicated that anti-trx-s lowered overall metabolic activities of mature seeds eliminating pre-harvest sprouting potential. Post-harvest ripening reactivated the metabolic activities of transgenic seeds to restore their germination vigor. These findings provided essential molecular phenomic information for PHS resistance of anti-trx-s and a credible strategy for future developing PHS resistant crops.
Collapse
Affiliation(s)
- Caixiang Liu
- CAS Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Centre for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, the Chinese Academy of Sciences, Wuhan 430071, China
| | - Feng Ding
- College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Fuhua Hao
- CAS Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Centre for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, the Chinese Academy of Sciences, Wuhan 430071, China
| | - Men Yu
- CAS Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Centre for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, the Chinese Academy of Sciences, Wuhan 430071, China.,Wuhan Zhongke Metaboss Ltd, 128 Guang-Gu-Qi-Lu, Wuhan 430074, China
| | - Hehua Lei
- CAS Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Centre for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, the Chinese Academy of Sciences, Wuhan 430071, China
| | - Xiangyu Wu
- CAS Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Centre for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, the Chinese Academy of Sciences, Wuhan 430071, China
| | - Zhengxi Zhao
- College of Plant Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Hongxiang Guo
- National Engineering Research Center for Wheat, Henan Agricultural University, Zhengzhou 450002, China
| | - Jun Yin
- National Engineering Research Center for Wheat, Henan Agricultural University, Zhengzhou 450002, China
| | - Yulan Wang
- CAS Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Centre for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, the Chinese Academy of Sciences, Wuhan 430071, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University, Hangzhou 310058, China
| | - Huiru Tang
- CAS Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Centre for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, the Chinese Academy of Sciences, Wuhan 430071, China.,State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Developmental Biology, Metabonomics and Systems Biology Laboratory, School of Life Sciences, Fudan University, Shanghai 200438, China
| |
Collapse
|