1
|
Gonciarz W, Kozlowska L, Róg J, Chmiela M. Untargeted metabolomic profiling for identifying systemic signatures of helicobacter pylori infection in a guinea pig model. Sci Rep 2025; 15:12889. [PMID: 40234702 PMCID: PMC12000522 DOI: 10.1038/s41598-025-98016-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 04/08/2025] [Indexed: 04/17/2025] Open
Abstract
Infections caused by the Gram-negative bacterium Helicobacter pylori (H. pylori) can lead to gastritis, gastric or duodenal ulcers, and even gastric cancer in humans. Investigating quantitative changes in soluble biomarkers associated with H. pylori infection offers a promising method for monitoring the progression of the infection, inflammatory response and potentially systemic consequences. This study aimed to identify, using an experimental model of H. pylori infection in guinea pigs, the specific metabolomic biomarkers in the serum of H. pylori-infected (32) versus uninfected (32) animals. The H. pylori status was confirmed through histological, molecular, and serological examinations. Metabolomic profiling was conducted using UPLC-QTOF/MS methods. The metabolomic biomarkers significantly associated with H. pylori infection were selected based on volcano plots and traditional univariate receiver operating characteristics (ROC). This study identified 12 unique metabolites significantly differentiating H. pylori-infected guinea pigs from uninfected ones. In summary, the metabolomic profiling of serum samples, in combination with ROC characteristics of the data, enhances the monitoring of H. pylori infection and related inflammatory responses in guinea pigs experimentally infected with these bacteria, with potential applications in humans for prediction the infection course and its systemic effects.
Collapse
Affiliation(s)
- Weronika Gonciarz
- Department of Immunology and Infectious Biology, Faculty of Biology and Environmental Protection, University of Lodz, 12/16 Banacha St., 90‑237, Lodz, Poland.
| | - Lucyna Kozlowska
- Laboratory of Human Metabolism Research, Department of Dietetics, Institute of Human Nutrition Sciences, Warsaw University of Life Sciences, 02-776, Warsaw, Poland
| | - Joanna Róg
- Laboratory of Human Metabolism Research, Department of Dietetics, Institute of Human Nutrition Sciences, Warsaw University of Life Sciences, 02-776, Warsaw, Poland
| | - Magdalena Chmiela
- Department of Immunology and Infectious Biology, Faculty of Biology and Environmental Protection, University of Lodz, 12/16 Banacha St., 90‑237, Lodz, Poland
| |
Collapse
|
2
|
Zhen LL, Feng L, Jiang WD, Wu P, Liu Y, Tang L, Li SW, Zhong CB, Zhou XQ. Exploring the novel benefits of leucine: Protecting nitrite-induced liver damage in sub-adult grass carp (Ctenopharyngodon idella) through regulating mitochondria quality control. FISH & SHELLFISH IMMUNOLOGY 2024; 151:109690. [PMID: 38866347 DOI: 10.1016/j.fsi.2024.109690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 05/21/2024] [Accepted: 06/10/2024] [Indexed: 06/14/2024]
Abstract
Leucine is an essential amino acid for fish. The ability of leucine to resist stress in fish has not been reported. Nitrite is a common pollutant in the aquatic environment. Therefore, we investigated the effects of dietary leucine on growth performance and nitrite-induced liver damage, mitochondrial dysfunction, autophagy, and apoptosis for sub-adult grass carp. A total of 450 grass carp (615.91 ± 1.15 g) were selected and randomly placed into 18 net cages. The leucine contents of the six diets were 2.91, 5.90, 8.92, 11.91, 14.93, and 17.92 g/kg, respectively. After a 9-week feeding trial, the nitrite exposure experiment was set up for 96 h. These results indicated that dietary leucine significantly promoted FW, WG, PWG, and SGR of sub-adult grass carp (P < 0.05). Appropriate levels of dietary leucine (11.91-17.92 g/kg) decreased the activities of serum parameters (glucose, cortisol, and methemoglobin contents, glutamic oxaloacetic transaminase, glutamic pyruvic transaminase, and lactate dehydrogenase), the contents of reactive oxygen species (ROS), nitric oxide (NO) and peroxynitrite (ONOO-). In addition, appropriate levels of dietary leucine (11.91-17.92 g/kg) increased the mRNA levels of mitochondrial biogenesis genes (PGC-1α, Nrf1/2, TFAM), fusion-related genes (Opa1, Mfn1/2) (P < 0.05), and decreased the mRNA levels of caspase 3, caspase 8, caspase 9, fission-related gene (Drp1), mitophagy-related genes (Pink1, Parkin) and autophagy-related genes (Beclin1, Ulk1, Atg5, Atg7, Atg12) (P < 0.05). Appropriate levels of dietary leucine (8.92-17.92 g/kg) also increased the protein levels of AMP-activated protein kinase (AMPK), prostacyclin (p62) and decreased the protein levels of protein light chain 3 (LC3), E3 ubiquitin ligase (Parkin), and Cytochrome c (Cytc). Appropriate levels of leucine (8.92-17.92 g/kg) could promote growth performance and alleviate nitrite-induced mitochondrial dysfunction, autophagy, apoptosis for sub-adult grass carp. Based on quadratic regression analysis of PWG and serum GPT activity, dietary leucine requirements of sub-adult grass carp were recommended to be 12.47 g/kg diet and 12.55 g/kg diet, respectively.
Collapse
Affiliation(s)
- Lu-Lu Zhen
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, China
| | - Lin Feng
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China; Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan, 611130, China
| | - Wei-Dan Jiang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China; Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan, 611130, China
| | - Pei Wu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China; Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan, 611130, China
| | - Yang Liu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China; Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan, 611130, China
| | - Ling Tang
- Animal Nutrition Institute, Sichuan Academy of Animal Science, Sichuan Animtech Feed Co. Ltd, Chengdu, 610066, Sichuan, China
| | - Shu-Wei Li
- Animal Nutrition Institute, Sichuan Academy of Animal Science, Sichuan Animtech Feed Co. Ltd, Chengdu, 610066, Sichuan, China
| | - Cheng-Bo Zhong
- Animal Nutrition Institute, Sichuan Academy of Animal Science, Sichuan Animtech Feed Co. Ltd, Chengdu, 610066, Sichuan, China
| | - Xiao-Qiu Zhou
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China; Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan, 611130, China.
| |
Collapse
|
3
|
Dziedzic M, Józefczuk E, Guzik TJ, Siedlinski M. Interplay Between Plasma Glycine and Branched-Chain Amino Acids Contributes to the Development of Hypertension and Coronary Heart Disease. Hypertension 2024; 81:1320-1331. [PMID: 38587181 PMCID: PMC11095885 DOI: 10.1161/hypertensionaha.123.22649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 03/28/2024] [Indexed: 04/09/2024]
Abstract
BACKGROUND Higher levels of plasma glycine are linked to a reduced risk, while increased levels of total branched-chain amino acids (tBCAAs) are associated with a higher risk of essential hypertension and coronary heart disease (CHD). As these metabolic components are interconnected, analyzing the tBCAAs/glycine ratio may help to understand their interplay in the pathogenesis of cardiovascular disease. METHODS The Cox regression approach was combined with the development of novel genetic tools for assessments of associations between plasma metabolomic data (glycine, tBCAAs, and tBCAAs/glycine ratio) from the UK Biobank and the development of hypertension and CHD. Genome-wide association study was performed on 186 523 White UK Biobank participants to identify new independent genetic instruments for the 2-sample Mendelian randomization analyses. P-gain statistic >10 identified instruments associated with tBCAAs/glycine ratio significantly stronger compared with individual amino acids. Outcomes of genome-wide association study on hypertension and CHD were derived from the UK Biobank (nonoverlapping sample), FinnGen, and CARDIoGRAMplusC4D. RESULTS The tBCAAs/glycine ratio was prospectively associated with a higher risk of developing hypertension and CHD (hazard ratio quintile Q5 versus Q1, 1.196 [95% CI, 1.109-1.289] and 1.226 [95% CI, 1.160-1.296], respectively). Mendelian randomization analysis demonstrated that tBCAAs/glycine ratio (P-gain >10) was a risk factor for hypertension (meta-analyzed inverse-variance weighted causal estimate 0.45 log odds ratio/SD (95% CI, 0.26-0.64) and CHD (0.48 [95% CI, 0.29-0.67]) with an absolute effect significantly larger compared with the effect of glycine (-0.06 [95% CI, -0.1 to -0.03] and -0.08 [95% CI, -0.11 to -0.05], respectively) or tBCAAs (0.22 [95% CI, 0.09-0.34] and 0.12 [95% CI, 0.01-0.24], respectively). CONCLUSIONS The total BCAAs/glycine ratio is a key element of the metabolic signature contributing to hypertension and CHD, which may reflect biological pathways shared by glycine and tBCAAs.
Collapse
Affiliation(s)
- Mateusz Dziedzic
- Department of Internal Medicine (M.D., E.J., T.J.G., M.S.), Faculty of Medicine, Jagiellonian University Medical College, Cracow, Poland
| | - Ewelina Józefczuk
- Department of Internal Medicine (M.D., E.J., T.J.G., M.S.), Faculty of Medicine, Jagiellonian University Medical College, Cracow, Poland
- Center for Medical Genomics OMICRON (T.J.G., M.S.), Faculty of Medicine, Jagiellonian University Medical College, Cracow, Poland
| | - Tomasz J. Guzik
- Department of Internal Medicine (M.D., E.J., T.J.G., M.S.), Faculty of Medicine, Jagiellonian University Medical College, Cracow, Poland
- Centre for Cardiovascular Science, Queen’s Medical Research Institute, University of Edinburgh, United Kingdom (T.J.G., M.S.)
| | - Mateusz Siedlinski
- Department of Internal Medicine (M.D., E.J., T.J.G., M.S.), Faculty of Medicine, Jagiellonian University Medical College, Cracow, Poland
- Center for Medical Genomics OMICRON (T.J.G., M.S.), Faculty of Medicine, Jagiellonian University Medical College, Cracow, Poland
- Centre for Cardiovascular Science, Queen’s Medical Research Institute, University of Edinburgh, United Kingdom (T.J.G., M.S.)
| |
Collapse
|
4
|
李 雨, 王 瑗, 袁 泉. [Latest Findings on the Role of α-Ketoglutarate in Metabolic Syndrome]. SICHUAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF SICHUAN UNIVERSITY. MEDICAL SCIENCE EDITION 2024; 55:783-792. [PMID: 38948289 PMCID: PMC11211801 DOI: 10.12182/20240560302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Indexed: 07/02/2024]
Abstract
Alpha-ketoglutarate (α-KG), an endogenous intermediate of the tricarboxylic acid cycle, is involved in a variety of cellular metabolic pathways. It serves as an energy donor, a precursor of amino acid biosynthesis, and an epigenetic regulator. α-KG plays physiological functions in immune regulation, oxidative stress, and anti-aging as well. In recent years, it has been reported that the level of α-KG in the body is closely associated with metabolic syndrome, including obesity, hyperglycemia, and other pathological factors. Exogenous supplementation of α-KG improves obesity, blood glucose levels, and cardiovascular disease risks associated with metabolic syndrome. Furthermore, α-KG regulates the common pathological mechanisms of metabolic syndrome, suggesting the potential application prospect of α-KG in metabolic syndrome. In order to provide a theoretical basis for further exploration of the application of α-KG in metabolic syndrome, we focused on α-KG and metabolic syndrome in this article and summarized the latest research progress in the role of α-KG in improving the pathological condition and disease progression of metabolic syndrome. For the next step, researchers may focus on the co-pathogenesis of metabolic syndrome and investigate whether α-KG can be used to achieve the therapeutic goal of "homotherapy for heteropathy" in the treatment of metabolic syndrome.
Collapse
Affiliation(s)
- 雨含 李
- 口腔疾病防治全国重点实验室 国家口腔医学中心 国家口腔疾病临床医学研究中心 四川大学华西口腔医院 种植科 (成都 610041)State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Dental Implant, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - 瑗 王
- 口腔疾病防治全国重点实验室 国家口腔医学中心 国家口腔疾病临床医学研究中心 四川大学华西口腔医院 种植科 (成都 610041)State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Dental Implant, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - 泉 袁
- 口腔疾病防治全国重点实验室 国家口腔医学中心 国家口腔疾病临床医学研究中心 四川大学华西口腔医院 种植科 (成都 610041)State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Dental Implant, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| |
Collapse
|
5
|
Wang Z, Yin Y, Mu Y, Cui L, Song X, Zhuang J, Gao S, Tao X, Chen W. Exploring the Occurrence Mechanism and Early-Warning Model of Phlebitis Induced by Aescinate Based on Metabolomics in Cerebral Infarction Patients. J Inflamm Res 2024; 17:343-355. [PMID: 38260811 PMCID: PMC10800284 DOI: 10.2147/jir.s436846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 12/28/2023] [Indexed: 01/24/2024] Open
Abstract
Objective This study aims to explore the mechanism underlying the induction of phlebitis by aescinate and create an early-warning model of phlebitis based on metabolomics. Methods Patients with cerebral infarction enrolled had been treated with aescinate. Plasma samples were collected either before administration of aescinate, upon the occurrence of phlebitis, or at the end of treatment. Non-targeted metabolomics and targeted amino acid metabolomics were carried out to analyze metabolic profiles and quantify the metabolites. Results Untargeted metabolomics revealed six differential metabolites in baseline samples versus post-treatment samples and four differential metabolites in baseline samples from patients with or without phlebitis. Pathways of these differential metabolites were mainly enriched in amino acid metabolism. Ten differential amino acids with a VIP value of >1 were identified in the baseline samples, enabling us to distinguish between patients with or without phlebitis. A logistic regression model was constructed (AUC 0.825) for early warning of phlebitis of grade 2 or higher. Conclusion The occurrence of aescinate-induced phlebitis, which can be predicted early during onset, may be associated with perturbations of the endogenous metabolic profile, especially the metabolism of amino acids.
Collapse
Affiliation(s)
- Zhipeng Wang
- Department of Pharmacy, Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), Shanghai, 200003, People’s Republic of China
| | - You Yin
- Department of Neurology, Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), Shanghai, 200003, People’s Republic of China
| | - Yuhui Mu
- Department of Pharmacy, Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), Shanghai, 200003, People’s Republic of China
- College of Traditional Chinese Medicine, Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan, 650500, People’s Republic of China
| | - Lili Cui
- Department of Pharmacy, Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), Shanghai, 200003, People’s Republic of China
| | - Xinhua Song
- Department of Pharmacy, Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), Shanghai, 200003, People’s Republic of China
| | - Jianhua Zhuang
- Department of Neurology, Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), Shanghai, 200003, People’s Republic of China
| | - Shouhong Gao
- Department of Pharmacy, Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), Shanghai, 200003, People’s Republic of China
| | - Xia Tao
- Department of Pharmacy, Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), Shanghai, 200003, People’s Republic of China
| | - Wansheng Chen
- Department of Pharmacy, Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), Shanghai, 200003, People’s Republic of China
| |
Collapse
|
6
|
Tekwe CD, Luan Y, Meininger CJ, Bazer FW, Wu G. Dietary supplementation with L-leucine reduces nitric oxide synthesis by endothelial cells of rats. Exp Biol Med (Maywood) 2023; 248:1537-1549. [PMID: 37837386 PMCID: PMC10676130 DOI: 10.1177/15353702231199078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Accepted: 07/21/2023] [Indexed: 10/16/2023] Open
Abstract
This study tested the hypothesis that elevated L-leucine concentrations in plasma reduce nitric oxide (NO) synthesis by endothelial cells (ECs) and affect adiposity in obese rats. Beginning at four weeks of age, male Sprague-Dawley rats were fed a casein-based low-fat (LF) or high-fat (HF) diet for 15 weeks. Thereafter, rats in the LF and HF groups were assigned randomly into one of two subgroups (n = 8/subgroup) and received drinking water containing either 1.02% L-alanine (isonitrogenous control) or 1.5% L-leucine for 12 weeks. The energy expenditure of the rats was determined at weeks 0, 6, and 11 of the supplementation period. At the end of the study, an oral glucose tolerance test was performed on all the rats immediately before being euthanized for the collection of tissues. HF feeding reduced (P < 0.001) NO synthesis in ECs by 21% and whole-body insulin sensitivity by 19% but increased (P < 0.001) glutamine:fructose-6-phosphate transaminase (GFAT) activity in ECs by 42%. Oral administration of L-leucine decreased (P < 0.05) NO synthesis in ECs by 14%, increased (P < 0.05) GFAT activity in ECs by 35%, and reduced (P < 0.05) whole-body insulin sensitivity by 14% in rats fed the LF diet but had no effect (P > 0.05) on these variables in rats fed the HF diet. L-Leucine supplementation did not affect (P > 0.05) weight gain, tissue masses (including white adipose tissue, brown adipose tissue, and skeletal muscle), or antioxidative capacity (indicated by ratios of glutathione/glutathione disulfide) in LF- or HF-fed rats and did not worsen (P > 0.05) adiposity, whole-body insulin sensitivity, or metabolic profiles in the plasma of obese rats. These results indicate that high concentrations of L-leucine promote glucosamine synthesis and impair NO production by ECs, possibly contributing to an increased risk of cardiovascular disease in diet-induced obese rats.
Collapse
Affiliation(s)
- Carmen D Tekwe
- Department of Animal Science, Texas A&M University, College Station, TX 77843, USA
- Department of Epidemiology and Biostatistics, Texas A&M University, College Station, TX 77843, USA
- Department of Epidemiology and Biostatistics, School of Public Health, Indiana University, Bloomington, IN 47403, USA
| | - Yuanyuan Luan
- Department of Epidemiology and Biostatistics, Texas A&M University, College Station, TX 77843, USA
- Department of Epidemiology and Biostatistics, School of Public Health, Indiana University, Bloomington, IN 47403, USA
| | - Cynthia J Meininger
- Department of Medical Physiology, Texas A&M University, College Station, TX 77843, USA
| | - Fuller W Bazer
- Department of Animal Science, Texas A&M University, College Station, TX 77843, USA
| | - Guoyao Wu
- Department of Animal Science, Texas A&M University, College Station, TX 77843, USA
- Department of Medical Physiology, Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
7
|
Mendelian Randomization Analysis Provides Insights into the Pathogenesis of Serum Levels of Branched-Chain Amino Acids in Cardiovascular Disease. Metabolites 2023; 13:metabo13030403. [PMID: 36984843 PMCID: PMC10059809 DOI: 10.3390/metabo13030403] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/03/2023] [Accepted: 03/06/2023] [Indexed: 03/11/2023] Open
Abstract
Several observational studies have indicated an association between high serum levels of branched-chain amino acids (BCAAs) and an increased risk of cardiovascular disease (CVD). To assess whether theses associations reflect causality, we carried out two-sample Mendelian randomization (MR). Single-nucleotide polymorphisms (SNPs) associated with BCAA were evaluated in 10 studies, including 24,925 participants. The association between SNPs and coronary artery disease (CAD) were assessed using summary estimates from the CARDIoGRAMplusC4D consortium. Further MR analysis of BCAAs and seven CVD outcomes was performed. The BCAA-raising gene functions were also analyzed. MR analyses revealed a risk-increasing causal relationship between serum BCAA concentrations and CAD (odds ratio 1.08; 95% confidence interval (CI) 1.02–1.14), which was partly mediated by blood pressure and type 2 diabetes. BCAA also demonstrated a causal relationship with ischemic CVD events induced by plaque rupture and thrombosis (false discovery rate <0.05). Two BCAA-raising genes (MRL33 and CBLN1) were preferentially associated with myocardial infarction risk in the presence of atherosclerosis (p < 0.003). Functional analysis of the BCAA-raising genes suggested the causal involvement of two pathophysiological pathways, including glucose metabolism (PPM1K and TRMT61A) related to plaque progression, and the newly discovered neuroendocrine disorders regulating blood pressure (MRPL33, CBLN1, and C2orf16) related to plaque rupture and thrombosis. This comprehensive MR analysis provided insights into the potential causal mechanisms linking BCAA with CVD risk and suggested targeting neuroendocrine disorders as a potential strategy for the prevention of CVD. These results warrant further studies to elucidate the mechanisms underlying these reported causal associations.
Collapse
|
8
|
Dela Justina V, Dos Passos Júnior RR, Lima VV, Giachini FR. Evidence of Nitric Oxide Impairment During Hypertensive Pregnancies. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1428:99-125. [PMID: 37466771 DOI: 10.1007/978-3-031-32554-0_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2023]
Abstract
Hypertensive disorders of pregnancy complicate up to 10% of pregnancies worldwide, and they can be classified into (1) gestational hypertension, (2) preeclampsia, (3) chronic hypertension and (4) chronic hypertension with preeclampsia. Nitric oxide (NO) plays an essential role in the haemodynamic adaptations observed during pregnancy. It has been shown that the nitric oxide pathway's dysfunction during pregnancy is associated with placental- and vascular-related diseases such as hypertensive disorders of pregnancy. This review aims to present a brief definition of hypertensive disorders of pregnancy and physiological maternal cardiovascular adaptations during pregnancy. We also detail how NO signalling is altered in the (a) systemic vasculature, (b) uterine artery/spiral arteries, (c) implantation and (d) placenta of hypertensive disorders during pregnancy. We conclude by summarizing the anti-hypertensive therapy of hypertensive disorders of pregnancy as a specific management strategy.
Collapse
Affiliation(s)
- Vanessa Dela Justina
- Graduate Program in Biological Sciences, Federal University of Goiás, Goiânia, Brazil
| | - Rinaldo Rodrigues Dos Passos Júnior
- Graduate Program in Biological Sciences, Federal University of Goiás, Goiânia, Brazil
- Institute of Health Sciences and Health, Universidad Federal De Mato Grosso, Barra do Garcas, Brazil
| | - Victor Vitorino Lima
- Institute of Health Sciences and Health, Universidad Federal De Mato Grosso, Barra do Garcas, Brazil
| | - Fernanda Regina Giachini
- Graduate Program in Biological Sciences, Federal University of Goiás, Goiânia, Brazil
- Institute of Health Sciences and Health, Universidad Federal De Mato Grosso, Barra do Garcas, Brazil
| |
Collapse
|
9
|
The Role of Amino Acids in Endothelial Biology and Function. Cells 2022; 11:cells11081372. [PMID: 35456051 PMCID: PMC9030017 DOI: 10.3390/cells11081372] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 04/08/2022] [Accepted: 04/16/2022] [Indexed: 12/20/2022] Open
Abstract
The vascular endothelium acts as an important component of the vascular system. It is a barrier between the blood and vessel wall. It plays an important role in regulating blood vessel tone, permeability, angiogenesis, and platelet functions. Several studies have shown that amino acids (AA) are key regulators in maintaining vascular homeostasis by modulating endothelial cell (EC) proliferation, migration, survival, and function. This review summarizes the metabolic and signaling pathways of AAs in ECs and discusses the importance of AA homeostasis in the functioning of ECs and vascular homeostasis. It also discusses the challenges in understanding the role of AA in the development of cardiovascular pathophysiology and possible directions for future research.
Collapse
|
10
|
Cui J, Shi Y, Xu X, Zhao F, Zhang J, Wei B. Identifying the cardioprotective mechanism of Danyu Tongmai Granules against myocardial infarction by targeted metabolomics combined with network pharmacology. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 98:153829. [PMID: 35104768 DOI: 10.1016/j.phymed.2021.153829] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 10/18/2021] [Accepted: 10/25/2021] [Indexed: 06/14/2023]
Abstract
BACKGROUND Danyu Tongmai Granules (DY), the commercial Chinese medicine, was well-accepted cardiovascular protective actions in clinic. However, the mechanisms underlying the beneficial effects of DY on cardiovascular disease still need largely to be clarified. PURPOSE Therefore, this study was designed to explore potential mechanisms of DY in myocardial infarction (MI) by integrated strategy of metabolomics and network pharmacology. METHODS Cardiomyocytes were subjected to H2O2 induced myocardial injury and rats were induced MI via isoproterenol (ISO) injection. The entire metabolic alterations in serum and heart tissues of experimental rats were profiled by UPLC-MS/MS. Based on the identified differential metabolites, the pathway analysis results were obtained and further validated using the network pharmacology approach. RESULTS We found that DY exerted significant cardioprotective effects in vitro and in vivo, and ameliorated inflammatory cell infiltration and cardiomyocyte apoptosis induced by ISO. The metabolomics data suggested that DY mainly affected the amino acid metabolism (i.e., valine, leucine and isoleucine biosynthesis, arginine biosynthesis, phenylalanine, tyrosine and tryptophan biosynthesis, phenylalanine metabolism, arginine biosynthesis, glycine, serine, as well as the alanine metabolism, aspartate and glutamate metabolism, etc.). Simultaneously, DY participated in the regulation of the biosynthesis of bile acids and biosynthesis of unsaturated fatty acids. Notably, DY significantly reduced the biosynthesis of valine, leucine and isoleucine to regulating the metabolism of branched chain amino acids (BCAAs) in infarcted myocardium, thus blocking the inflammation via inhibiting the expression of NLRP3 inflammasome in ISO-induced rats. The anti-inflammatory system of DY was further validated with the results of network pharmacology. CONCLUSION Our study, for the first time, confirmed that DY inhibited inflammation and further exerted significant anti-myocardial infarction effect. Additionally, our work further demonstrated that the myocardial protective effect of DY was contributed to the inhibition of the NLRP3 inflammasome activation by regulating BCAAs in infarcted myocardium using the comprehensive metabolomics, molecular biology and network analysis. Overall, our study gained new insights into the role of the relationship between the metabolic regulation of BCAAs and the NLRP3 inflammasome against MI.
Collapse
Affiliation(s)
- Jing Cui
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, No. 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Yangyang Shi
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, No. 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Xueli Xu
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, No. 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Fei Zhao
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, No. 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Ji Zhang
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China; Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou 450052, China
| | - Bo Wei
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, No. 100 Kexue Avenue, Zhengzhou, Henan 450001, China.
| |
Collapse
|
11
|
Wang R, Xie L, Zhang J, Li J, Xie H, Yang Y, Ren H, Shang Z, Liu C. Characterization of the Metabolites and Construction of a Novel Diagnostic Panel in Calcium Oxalate Urolithiasis by Electrospray Ionization – Mass Spectrometry (ESI-MS) Metabolomics. ANAL LETT 2022. [DOI: 10.1080/00032719.2022.2040523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Affiliation(s)
- Rui Wang
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Linguo Xie
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Jingdong Zhang
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Jie Li
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Haijie Xie
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Yu Yang
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Haotian Ren
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Zhiqun Shang
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Chunyu Liu
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| |
Collapse
|
12
|
Price D, Jackson KG, Lovegrove JA, Givens DI. The effects of whey proteins, their peptides and amino acids on vascular function. NUTR BULL 2022; 47:9-26. [DOI: 10.1111/nbu.12543] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 01/12/2022] [Accepted: 01/12/2022] [Indexed: 12/14/2022]
Affiliation(s)
- Drew Price
- Hugh Sinclair Unit of Human Nutrition Department of Food and Nutritional Sciences University of Reading Reading UK
- Institute of Food Nutrition and Health University of Reading Reading UK
| | - Kim G. Jackson
- Hugh Sinclair Unit of Human Nutrition Department of Food and Nutritional Sciences University of Reading Reading UK
- Institute of Food Nutrition and Health University of Reading Reading UK
- Institute of Cardiovascular and Metabolic Research University of Reading Reading UK
| | - Julie A. Lovegrove
- Hugh Sinclair Unit of Human Nutrition Department of Food and Nutritional Sciences University of Reading Reading UK
- Institute of Food Nutrition and Health University of Reading Reading UK
- Institute of Cardiovascular and Metabolic Research University of Reading Reading UK
| | - David Ian Givens
- Institute of Food Nutrition and Health University of Reading Reading UK
| |
Collapse
|
13
|
Ding Y, Zhou Y, Ling P, Feng X, Luo S, Zheng X, Little PJ, Xu S, Weng J. Metformin in cardiovascular diabetology: a focused review of its impact on endothelial function. Am J Cancer Res 2021; 11:9376-9396. [PMID: 34646376 PMCID: PMC8490502 DOI: 10.7150/thno.64706] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 08/30/2021] [Indexed: 02/07/2023] Open
Abstract
As a first-line treatment for diabetes, the insulin-sensitizing biguanide, metformin, regulates glucose levels and positively affects cardiovascular function in patients with diabetes and cardiovascular complications. Endothelial dysfunction (ED) represents the primary pathological change of multiple vascular diseases, because it causes decreased arterial plasticity, increased vascular resistance, reduced tissue perfusion and atherosclerosis. Caused by “biochemical injury”, ED is also an independent predictor of cardiovascular events. Accumulating evidence shows that metformin improves ED through liver kinase B1 (LKB1)/5'-adenosine monophosphat-activated protein kinase (AMPK) and AMPK-independent targets, including nuclear factor-kappa B (NF-κB), phosphatidylinositol 3 kinase-protein kinase B (PI3K-Akt), endothelial nitric oxide synthase (eNOS), sirtuin 1 (SIRT1), forkhead box O1 (FOXO1), krüppel-like factor 4 (KLF4) and krüppel-like factor 2 (KLF2). Evaluating the effects of metformin on endothelial cell functions would facilitate our understanding of the therapeutic potential of metformin in cardiovascular diabetology (including diabetes and its cardiovascular complications). This article reviews the physiological and pathological functions of endothelial cells and the intact endothelium, reviews the latest research of metformin in the treatment of diabetes and related cardiovascular complications, and focuses on the mechanism of action of metformin in regulating endothelial cell functions.
Collapse
|
14
|
Zheng J, Zheng C, Song B, Guo Q, Zhong Y, Zhang S, Zhang L, Duan G, Li F, Duan Y. HMB Improves Lipid Metabolism of Bama Xiang Mini-Pigs via Modulating the Bacteroidetes-Acetic Acid-AMPKα Axis. Front Microbiol 2021; 12:736997. [PMID: 34484171 PMCID: PMC8415715 DOI: 10.3389/fmicb.2021.736997] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 07/28/2021] [Indexed: 12/29/2022] Open
Abstract
Here, we used Bama Xiang mini-pigs to explore the effects of different dietary β-hydroxy-β-methylbutyrate (HMB) levels (0, 0.13, 0.64 or 1.28%) on lipid metabolism of adipose tissue. Results showed that HMB decreased the fat percentage of pigs (linearly, P < 0.05), and the lowest value was observed in the 0.13% HMB group. Moreover, the colonic acetic acid concentration and the relative Bacteroidetes abundance were increased in response to HMB supplementation (P < 0.05). Correlation analysis identified a positive correlation between the relative Bacteroidetes abundance and acetic acid production, and a negative correlation between fat percentage and the relative Bacteroidetes abundance or acetic acid production. HMB also upregulated the phosphorylation (p) of AMPKα, Sirt1, and FoxO1, and downregulated the p-mTOR expression. Collectively, these findings indicate that reduced fat percentage in Bama Xiang mini-pigs could be induced by HMB supplementation and the mechanism might be associated with the Bacteroidetes-acetic acid-AMPKα axis.
Collapse
Affiliation(s)
- Jie Zheng
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China.,College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Changbing Zheng
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
| | - Bo Song
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China.,College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Qiuping Guo
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
| | - Yinzhao Zhong
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
| | - Shiyu Zhang
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
| | - Lingyu Zhang
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China.,College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Geyan Duan
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China.,College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Fengna Li
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
| | - Yehui Duan
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
| |
Collapse
|
15
|
Moslehi N, Mirmiran P, Marzbani R, Rezadoost H, Mirzaie M, Azizi F, Tehrani FR. Serum metabolomics study of women with different annual decline rates of anti-Müllerian hormone: an untargeted gas chromatography-mass spectrometry-based study. Hum Reprod 2021; 36:721-733. [PMID: 33320198 DOI: 10.1093/humrep/deaa279] [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] [Received: 05/13/2020] [Revised: 09/02/2020] [Indexed: 01/09/2023] Open
Abstract
STUDY QUESTION Which metabolites are associated with varying rates of ovarian aging, measured as annual decline rates of anti-Müllerian hormone (AMH) concentrations? SUMMARY ANSWER Higher serum concentrations of metabolites of phosphate, N-acetyl-d-glucosamine, branched chained amino acids (BCAAs), proline, urea and pyroglutamic acid were associated with higher odds of fast annual decline rate of AMH. WHAT IS KNOWN ALREADY Age-related rate of ovarian follicular loss varies among women, and the factors underlying such inter-individual variations are mainly unknown. The rate of ovarian aging is clinically important due to its effects on both reproduction and health of women. Metabolomics, a global investigation of metabolites in biological samples, provides an opportunity to study metabolites or metabolic pathways in relation to a physiological/pathophysiological condition. To date, no metabolomics study has been conducted regarding the differences in the rates of ovarian follicular loss. STUDY DESIGN, SIZE, DURATION This prospective study was conducted on 186 reproductive-aged women with regular menstrual cycles and history of natural fertility, randomly selected using random case selection option in SPSS from the Tehran Lipid and Glucose Study. PARTICIPANTS/MATERIALS, SETTING, METHODS AMH concentrations were measured at baseline (1999-2001) and the fifth follow-up examination (2014-2017), after a median follow-up of 16 years, by immunoassay using Gen II kit. The annual decline rate of AMH was calculated by dividing the AMH decline rate by the follow-up duration (percent/year). The women were categorized based on the tertiles of the annual decline rates. Untargeted metabolomics analysis of the fasting-serum samples collected during the second follow-up examination cycle (2005-2008) was performed using gas chromatography-mass spectrometry. A combination of univariate and multivariate approaches was used to investigate the associations between metabolites and the annual decline rates of AMH. MAIN RESULTS AND THE ROLE OF CHANCE After adjusting the baseline values of age, AMH and BMI, 29 metabolites were positively correlated with the annual AMH decline rates. The comparisons among the tertiles of the annual decline rate of AMH revealed an increase in the relative abundance of 15 metabolites in the women with a fast decline (tertile 3), compared to those with a slow decline (tertile 1). There was no distinct separation between women with slow and fast decline rates while considering 41 metabolites simultaneously using the principal component analysis and the partial least-squares discriminant analysis models. The odds of fast AMH decline was increased with higher serum metabolites of phosphate, N-acetyl-d-glucosamine, BCAAs, proline, urea and pyroglutamic acid. Amino sugar and nucleotide sugar metabolism, BCAAs metabolism and aminoacyl tRNA biosynthesis were among the most significant pathways associated with the fast decline rate of AMH. LIMITATIONS, REASONS FOR CAUTION Estimating the annual decline rates of AMH using the only two measures of AMH is the main limitation of the study which assumes a linear fixed reduction in AMH during the study. Since using the two-time points did not account for the variability in the decline rate of AMH, the annual decline rates estimated in this study may not accurately show the trend of the reduction in AMH. In addition, despite the longitudinal nature of the study and statistical adjustment of the participants' ages, it is difficult to distinguish the AMH-related metabolites observed in this study can accelerate ovarian aging or they are reflections of different rates of the aging process. WIDER IMPLICATIONS OF THE FINDINGS Some metabolite features related to the decline rates of AMH have been suggested in this study; further prospective studies with multiple measurements of AMH are needed to confirm the findings of this study and to better understand the molecular process underlying variations in ovarian aging. STUDY FUNDING/COMPETING INTEREST(S) This study, as a part of PhD thesis of Ms Nazanin Moslehi, was supported by Shahid Beheshti University of Medical Sciences (10522-4). There were no competing interests. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- Nazanin Moslehi
- Nutrition and Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Parvin Mirmiran
- Nutrition and Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Department of Clinical Nutrition and Dietetics, Faculty of Nutrition and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Rezvan Marzbani
- Department of Phytochemistry, Medicinal Plants and Drugs Research Institute, Shahid Beheshti University, Tehran, Iran
| | - Hassan Rezadoost
- Department of Phytochemistry, Medicinal Plants and Drugs Research Institute, Shahid Beheshti University, Tehran, Iran
| | - Mehdi Mirzaie
- Department of Applied Mathematics, Faculty of Mathematical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Fereidoun Azizi
- Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fahimeh Ramezani Tehrani
- Reproductive Endocrinology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
16
|
Mahbub MH, Yamaguchi N, Hase R, Takahashi H, Ishimaru Y, Watanabe R, Saito H, Shimokawa J, Yamamoto H, Kikuchi S, Tanabe T. Plasma Branched-Chain and Aromatic Amino Acids in Relation to Hypertension. Nutrients 2020; 12:nu12123791. [PMID: 33322015 PMCID: PMC7764357 DOI: 10.3390/nu12123791] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 12/07/2020] [Accepted: 12/09/2020] [Indexed: 12/15/2022] Open
Abstract
Findings of the available studies regarding the roles of branched-chain amino acids (BCAAs) and aromatic amino acids (AAAs) in hypertension are inconsistent, conflicting and inconclusive. The purpose of this study was to explore and clarify the existence of any relationships of individual BCAAs and AAAs with hypertension with adjustments for potential relevant confounders. A total of 2805 healthy controls and 2736 hypertensive patients were included in the current analysis. The associations between individual amino acids and hypertension were explored by logistic regression analyses adjusted for potential confounding variables. Among the investigated amino acids, only the BCAAs showed consistently significant positive associations with hypertension in the adjusted models (p-trend < 0.05 to 0.001). However, compared with the corresponding lowest quartile of individual BCAAs, the positive association with hypertension remained significant only in the highest quartile (p < 0.01 to 0.001). We confirmed in a relatively large cohort of subjects that BCAAs, not AAAs, demonstrated consistent positive associations with hypertension. The results display the promising potential for the use of BCAAs as relevant and accessible biomarkers, and provide perspectives on interventions directed towards the reduction in plasma BCAA levels in the prevention and management of hypertension.
Collapse
Affiliation(s)
- M. H. Mahbub
- Department of Public Health and Preventive Medicine, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi 755-8505, Japan; (N.Y.); (R.H.); (Y.I.); (R.W.); (H.S.); (J.S.); (T.T.)
- Correspondence: ; Tel.: +81-836-22-2231
| | - Natsu Yamaguchi
- Department of Public Health and Preventive Medicine, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi 755-8505, Japan; (N.Y.); (R.H.); (Y.I.); (R.W.); (H.S.); (J.S.); (T.T.)
| | - Ryosuke Hase
- Department of Public Health and Preventive Medicine, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi 755-8505, Japan; (N.Y.); (R.H.); (Y.I.); (R.W.); (H.S.); (J.S.); (T.T.)
| | - Hidekazu Takahashi
- Department of Public Health, Faculty of Veterinary Medicine, Okayama University of Science, Imabari, Ehime 794-8555, Japan;
| | - Yasutaka Ishimaru
- Department of Public Health and Preventive Medicine, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi 755-8505, Japan; (N.Y.); (R.H.); (Y.I.); (R.W.); (H.S.); (J.S.); (T.T.)
| | - Rie Watanabe
- Department of Public Health and Preventive Medicine, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi 755-8505, Japan; (N.Y.); (R.H.); (Y.I.); (R.W.); (H.S.); (J.S.); (T.T.)
| | - Hiroyuki Saito
- Department of Public Health and Preventive Medicine, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi 755-8505, Japan; (N.Y.); (R.H.); (Y.I.); (R.W.); (H.S.); (J.S.); (T.T.)
| | - Junki Shimokawa
- Department of Public Health and Preventive Medicine, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi 755-8505, Japan; (N.Y.); (R.H.); (Y.I.); (R.W.); (H.S.); (J.S.); (T.T.)
| | - Hiroshi Yamamoto
- Institute for Innovation, Ajinomoto Co. Inc., Kawasaki, Kanagawa 210-8681, Japan; (H.Y.); (S.K.)
| | - Shinya Kikuchi
- Institute for Innovation, Ajinomoto Co. Inc., Kawasaki, Kanagawa 210-8681, Japan; (H.Y.); (S.K.)
| | - Tsuyoshi Tanabe
- Department of Public Health and Preventive Medicine, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi 755-8505, Japan; (N.Y.); (R.H.); (Y.I.); (R.W.); (H.S.); (J.S.); (T.T.)
| |
Collapse
|
17
|
Fedorova EN, Varlamova DO, Kivero AD, Guk KD, Ptitsyn LR. Ultra-performance liquid chromatography (UPLC) for the determination of organic acids – The intermediates of branched-chain amino acid biosynthesis in Escherichia coli strains. J LIQ CHROMATOGR R T 2020. [DOI: 10.1080/10826076.2020.1832894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
18
|
Reho JJ, Guo DF, Rahmouni K. Mechanistic Target of Rapamycin Complex 1 Signaling Modulates Vascular Endothelial Function Through Reactive Oxygen Species. J Am Heart Assoc 2020; 8:e010662. [PMID: 31020916 PMCID: PMC6512105 DOI: 10.1161/jaha.118.010662] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Background The mechanistic target of rapamycin complex 1 (mTORC1) is an important intracellular energy sensor that regulates gene expression and protein synthesis through its downstream signaling components, the S6‐kinase and the ribosomal S6 protein. Recently, signaling arising from mTORC1 has been implicated in regulation of the cardiovascular system with implications for disease. Here, we examined the contribution of mTORC1 signaling to the regulation of vascular function. Methods and Results Activation of mTORC1 pathway in aortic rings with leucine or an adenoviral vector expressing a constitutively active S6‐kinase reduces endothelial‐dependent vasorelaxation in an mTORC1‐dependent manner without affecting smooth muscle relaxation responses. Moreover, activation of mTORC1 signaling in endothelial cells increases reactive oxygen species (ROS) generation and ROS gene expression resulting in a pro‐oxidant gene environment. Blockade of ROS signaling with Tempol restores endothelial function in vascular rings with increased mTORC1 activity indicating a crucial interaction between mTORC1 and ROS signaling. We then tested the role of nuclear factor‐κB transcriptional complex in connecting mTORC1 and ROS signaling in endothelial cells. Blockade of inhibitor of nuclear factor κ‐B kinase subunit β activity with BMS‐345541 prevented the increased ROS generation associated with increased mTORC1 activity in endothelial cells but did not improve vascular endothelial function in aortic rings with increased mTORC1 and ROS signaling. Conclusions These results implicate mTORC1 as a critical molecular signaling hub in the vascular endothelium in mediating vascular endothelial function through modulation of ROS signaling.
Collapse
Affiliation(s)
- John J Reho
- 1 Department of Pharmacology University of Iowa Carver College of Medicine Iowa City IA
| | - Deng-Fu Guo
- 1 Department of Pharmacology University of Iowa Carver College of Medicine Iowa City IA
| | - Kamal Rahmouni
- 1 Department of Pharmacology University of Iowa Carver College of Medicine Iowa City IA.,2 Department of Internal Medicine University of Iowa Carver College of Medicine Iowa City IA.,3 Fraternal Order of Eagles Diabetes Research Center University of Iowa Iowa City IA
| |
Collapse
|
19
|
Tekwe CD, Yao K, Lei J, Li X, Gupta A, Luan Y, Meininger CJ, Bazer FW, Wu G. Oral administration of α-ketoglutarate enhances nitric oxide synthesis by endothelial cells and whole-body insulin sensitivity in diet-induced obese rats. Exp Biol Med (Maywood) 2019; 244:1081-1088. [PMID: 31357871 DOI: 10.1177/1535370219865229] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Obesity is a risk factor for many chronic diseases, including hypertension, type-2 diabetes, and cancer. Interestingly, concentrations of branched-chain amino acids (BCAAs) in plasma are commonly associated with endothelial dysfunction in humans and animals with obesity. Because L-leucine inhibits nitric oxide synthesis by endothelial cells (EC), we hypothesized that dietary supplementation with AKG (a substrate for BCAA transaminase) may stimulate BCAA catabolism in the small intestine and extra-intestinal tissues, thereby reducing the circulating concentrations of BCAAs and increasing nitric oxide synthesis by endothelial cells. Beginning at four weeks of age, male Sprague-Dawley rats were fed a low-fat or a high-fat diet for 15 weeks. At 19 weeks of age, lean or obese rats continued to be fed for 12 weeks their respective diets and received drinking water containing 0 or 1% AKG ( n = 8/group). At 31 weeks of age, the rats were euthanized to obtain tissues. Food intake did not differ ( P > 0.05) between rats supplemented with or without AKG. Oral administration of AKG (250 mg/kg BW per day) reduced ( P < 0.05) concentrations of BCAAs, glucose, ammonia, and triacylglycerols in plasma, adiposity, and glutamine:fructose-6-phosphate transaminase activity in endothelial cells, and enhanced ( P < 0.05) concentrations of the reduced form of glutathione in tissues, nitric oxide synthesis by endothelial cells, and whole-body insulin sensitivity (indicated by oral glucose tolerance test) in both low-fat and high-fat rats. AKG administration reduced ( P < 0.05) white adipose tissue weights of rats in the low-fat and high-fat groups. These novel results indicate that AKG can reduce adiposity and increase nitric oxide production by endothelial cells in diet-induced obese rats. Impact statement Obesity is associated with elevated concentrations of branched-chain amino acids, including L-leucine. L-Leucine inhibits the synthesis of nitric oxide from L-arginine by endothelial cells, contributing to impairments in angiogenesis, blood flow, and vascular dysfunction, as well as insulin resistance. Reduction in the circulating levels of branched-chain amino acids through dietary supplementation with α-ketoglutarate to promote their transamination in the small intestine and other tissues can restore nitric oxide synthesis in the vasculature and reduce the weights of white adipose tissues, thereby improving metabolic profiles and whole-body insulin sensitivity (indicated by oral glucose tolerance test) in diet-induced obese rats. Our findings provide a simple and effective nutritional means to alleviate metabolic syndrome in obese subjects. This is highly significant to combat the current obesity epidemic and associated health problems in humans worldwide.
Collapse
Affiliation(s)
- Carmen D Tekwe
- Department of Animal Science, Texas A&M University, College Station, TX 77843, USA.,Department of Epidemiology and Biostatistics, Texas A&M University, College Station, TX 77843, USA
| | - Kang Yao
- Department of Animal Science, Texas A&M University, College Station, TX 77843, USA
| | - Jian Lei
- Department of Animal Science, Texas A&M University, College Station, TX 77843, USA
| | - Xilong Li
- Department of Animal Science, Texas A&M University, College Station, TX 77843, USA
| | - Anand Gupta
- Department of Epidemiology and Biostatistics, Texas A&M University, College Station, TX 77843, USA
| | - Yuanyuan Luan
- Department of Epidemiology and Biostatistics, Texas A&M University, College Station, TX 77843, USA
| | - Cynthia J Meininger
- Department of Medical Physiology, Texas A&M Health Science Center, College Station, TX 77843, USA
| | - Fuller W Bazer
- Department of Animal Science, Texas A&M University, College Station, TX 77843, USA
| | - Guoyao Wu
- Department of Animal Science, Texas A&M University, College Station, TX 77843, USA.,Department of Medical Physiology, Texas A&M Health Science Center, College Station, TX 77843, USA
| |
Collapse
|
20
|
Wang YY, Xu JZ, Zhang WG. Metabolic engineering of l-leucine production in Escherichia coli and Corynebacterium glutamicum: a review. Crit Rev Biotechnol 2019; 39:633-647. [PMID: 31055970 DOI: 10.1080/07388551.2019.1577214] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
l-Leucine, as an essential branched-chain amino acid for humans and animals, has recently been attracting much attention because of its potential for a fast-growing market demand. The applicability ranges from flavor enhancers, animal feed additives and ingredients in cosmetic to specialty nutrients in pharmaceutical and medical fields. Microbial fermentation is the major method for producing l-leucine by using Escherichia coli and Corynebacterium glutamicum as host bacteria. This review gives an overview of the metabolic pathway of l-leucine (i.e. production, import and export systems) and highlights the main regulatory mechanisms of operons in E. coli and C. glutamicum l-leucine biosynthesis. We summarize here the current trends in metabolic engineering techniques and strategies for manipulating l-leucine producing strains. Finally, future perspectives to construct industrially advantageous strains are considered with respect to recent advances in biology.
Collapse
Affiliation(s)
- Ying-Yu Wang
- a The Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology , Jiangnan University , WuXi , People's Republic of China
| | - Jian-Zhong Xu
- a The Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology , Jiangnan University , WuXi , People's Republic of China.,b The Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology , Jiangnan University , WuXi , People's Republic of China
| | - Wei-Guo Zhang
- a The Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology , Jiangnan University , WuXi , People's Republic of China
| |
Collapse
|
21
|
Gallic Acid-L-Leucine Conjugate Protects Mice against LPS-Induced Inflammation and Sepsis via Correcting Proinflammatory Lipid Mediator Profiles and Oxidative Stress. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:1081287. [PMID: 29765489 PMCID: PMC5889890 DOI: 10.1155/2018/1081287] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2017] [Revised: 12/19/2017] [Accepted: 01/08/2018] [Indexed: 12/20/2022]
Abstract
The pathology of endotoxin LPS-induced sepsis is hallmarked by aberrant production of proinflammatory lipid mediators and nitric oxide (NO). The aim of the present study was to determine whether the new product gallic acid-L-leucine (GAL) conjugate could ameliorate the LPS-induced dysregulation of arachidonic acid metabolism and NO production. We first investigated the effects of GAL conjugate on the expression of proinflammatory enzymes and the production of proinflammatory NO and lipid mediators in mouse macrophage cell line RAW264.7, primary peritoneal macrophages, and mouse model. Western blot analyses revealed that GAL attenuated LPS-induced expression of iNOS, COX-2, and 5-LOX in a concentration-dependent manner. Consistently, probing NO-mediated fluorescence revealed that GAL antagonized the stimulatory effect of LPS on iNOS activity. By profiling of lipid mediators with ESI-MS-based lipidomics, we found that GAL suppressed LPS-induced overproduction of prostaglandin E2, prostaglandin F2, leukotriene B4, and thromboxane B2. We further discovered that GAL might exhibit anti-inflammatory activities by the following mechanisms: (1) suppressing LPS-induced activation of MAP kinases (i.e., ERK1/2, JNK, and p38); (2) reducing the production of reactive oxygen species (ROS); and (3) preventing LPS-induced nuclear translocation of transcription factors NF-κB and AP-1. Consequently, GAL significantly decreased the levels of COX-2 and iNOS expression and the plasma levels of proinflammatory lipid mediators in LPS-treated mice. GAL pretreatment enhanced the survival of mice against LPS-induced endotoxic shock. Taken together, our results suggest that GAL may be a potential anti-inflammatory drug for the treatment of endotoxemia and sepsis.
Collapse
|
22
|
Eelen G, de Zeeuw P, Treps L, Harjes U, Wong BW, Carmeliet P. Endothelial Cell Metabolism. Physiol Rev 2018; 98:3-58. [PMID: 29167330 PMCID: PMC5866357 DOI: 10.1152/physrev.00001.2017] [Citation(s) in RCA: 367] [Impact Index Per Article: 52.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 06/19/2017] [Accepted: 06/22/2017] [Indexed: 02/06/2023] Open
Abstract
Endothelial cells (ECs) are more than inert blood vessel lining material. Instead, they are active players in the formation of new blood vessels (angiogenesis) both in health and (life-threatening) diseases. Recently, a new concept arose by which EC metabolism drives angiogenesis in parallel to well-established angiogenic growth factors (e.g., vascular endothelial growth factor). 6-Phosphofructo-2-kinase/fructose-2,6-bisphosphatase-3-driven glycolysis generates energy to sustain competitive behavior of the ECs at the tip of a growing vessel sprout, whereas carnitine palmitoyltransferase 1a-controlled fatty acid oxidation regulates nucleotide synthesis and proliferation of ECs in the stalk of the sprout. To maintain vascular homeostasis, ECs rely on an intricate metabolic wiring characterized by intracellular compartmentalization, use metabolites for epigenetic regulation of EC subtype differentiation, crosstalk through metabolite release with other cell types, and exhibit EC subtype-specific metabolic traits. Importantly, maladaptation of EC metabolism contributes to vascular disorders, through EC dysfunction or excess angiogenesis, and presents new opportunities for anti-angiogenic strategies. Here we provide a comprehensive overview of established as well as newly uncovered aspects of EC metabolism.
Collapse
Affiliation(s)
- Guy Eelen
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven, Belgium; and Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Leuven, Belgium
| | - Pauline de Zeeuw
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven, Belgium; and Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Leuven, Belgium
| | - Lucas Treps
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven, Belgium; and Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Leuven, Belgium
| | - Ulrike Harjes
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven, Belgium; and Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Leuven, Belgium
| | - Brian W Wong
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven, Belgium; and Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Leuven, Belgium
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven, Belgium; and Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Leuven, Belgium
| |
Collapse
|
23
|
Association of plasma free amino acids with hyperuricemia in relation to diabetes mellitus, dyslipidemia, hypertension and metabolic syndrome. Sci Rep 2017; 7:17616. [PMID: 29247200 PMCID: PMC5732272 DOI: 10.1038/s41598-017-17710-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 11/29/2017] [Indexed: 12/22/2022] Open
Abstract
Previous studies demonstrated independent contributions of plasma free amino acids (PFAAs) and high uric acid (UA) concentrations to increased risks of lifestyle-related diseases (LSRDs), but the important associations between these factors and LSRDs remain unknown. We quantified PFAAs and UA amongst Japanese subjects without LSRDs (no-LSRD, n = 2805), and with diabetes mellitus (DM, n = 415), dyslipidemia (n = 3207), hypertension (n = 2736) and metabolic syndrome (MetS, n = 717). The concentrations of most amino acids differed significantly between the subjects with and without hyperuricemia (HU) and also between the no-LSRD and LSRD groups (p < 0.05 to 0.001). After adjustment, the logistic regression analyses revealed that lysine in DM, alanine, proline and tyrosine in dyslipidemia, histidine, lysine and ornithine in hypertension, and lysine and tyrosine in MetS demonstrated significant positive associations with HU among the patients with LSRDs only (p < 0.05 to 0.005). By contrast, arginine, asparagine and threonine showed significant inverse associations with HU in the no-LSRD group only (p < 0.05 to 0.01). For the first time, we provide evidence for distinct patterns of association between PFAAs and HU in LSRDs, and postulate the possibility of interplay between PFAAs and UA in their pathophysiology.
Collapse
|
24
|
Briggs MA, Petersen KS, Kris-Etherton PM. Saturated Fatty Acids and Cardiovascular Disease: Replacements for Saturated Fat to Reduce Cardiovascular Risk. Healthcare (Basel) 2017; 5:E29. [PMID: 28635680 PMCID: PMC5492032 DOI: 10.3390/healthcare5020029] [Citation(s) in RCA: 160] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 06/13/2017] [Accepted: 06/16/2017] [Indexed: 02/07/2023] Open
Abstract
Dietary recommendations to decrease the risk of cardiovascular disease (CVD) have focused on reducing intake of saturated fatty acids (SFA) for more than 50 years. While the 2015-2020 Dietary Guidelines for Americans advise substituting both monounsaturated and polyunsaturated fatty acids for SFA, evidence supports other nutrient substitutions that will also reduce CVD risk. For example, replacing SFA with whole grains, but not refined carbohydrates, reduces CVD risk. Replacing SFA with protein, especially plant protein, may also reduce CVD risk. While dairy fat (milk, cheese) is associated with a slightly lower CVD risk compared to meat, dairy fat results in a significantly greater CVD risk relative to unsaturated fatty acids. As research continues, we will refine our understanding of dietary patterns associated with lower CVD risk.
Collapse
Affiliation(s)
- Michelle A Briggs
- Department of Biology, Lycoming College, 700 College Place, Williamsport, PA 17701, USA.
| | - Kristina S Petersen
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA 16802, USA.
| | - Penny M Kris-Etherton
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA 16802, USA.
| |
Collapse
|
25
|
Gene-Diet Interactions in Type 2 Diabetes: The Chicken and Egg Debate. Int J Mol Sci 2017; 18:ijms18061188. [PMID: 28574454 PMCID: PMC5486011 DOI: 10.3390/ijms18061188] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 05/23/2017] [Accepted: 05/26/2017] [Indexed: 02/07/2023] Open
Abstract
Consistent evidence from both experimental and human studies indicates that Type 2 diabetes mellitus (T2DM) is a complex disease resulting from the interaction of genetic, epigenetic, environmental, and lifestyle factors. Nutrients and dietary patterns are important environmental factors to consider in the prevention, development and treatment of this disease. Nutritional genomics focuses on the interaction between bioactive food components and the genome and includes studies of nutrigenetics, nutrigenomics and epigenetic modifications caused by nutrients. There is evidence supporting the existence of nutrient-gene and T2DM interactions coming from animal studies and family-based intervention studies. Moreover, many case-control, cohort, cross-sectional cohort studies and clinical trials have identified relationships between individual genetic load, diet and T2DM. Some of these studies were on a large scale. In addition, studies with animal models and human observational studies, in different countries over periods of time, support a causative relationship between adverse nutritional conditions during in utero development, persistent epigenetic changes and T2DM. This review provides comprehensive information on the current state of nutrient-gene interactions and their role in T2DM pathogenesis, the relationship between individual genetic load and diet, and the importance of epigenetic factors in influencing gene expression and defining the individual risk of T2DM.
Collapse
|
26
|
Bifari F, Nisoli E. Branched-chain amino acids differently modulate catabolic and anabolic states in mammals: a pharmacological point of view. Br J Pharmacol 2017; 174:1366-1377. [PMID: 27638647 PMCID: PMC5429325 DOI: 10.1111/bph.13624] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Revised: 07/28/2016] [Accepted: 08/03/2016] [Indexed: 12/21/2022] Open
Abstract
Substantial evidence has been accumulated suggesting that branched-chain amino acid (BCAA) supplementation or BCAA-rich diets have a positive effect on the regulation of body weight, muscle protein synthesis, glucose homeostasis, the ageing process and extend healthspan. Despite these beneficial effects, epidemiological studies have shown that BCAA plasma concentrations and BCAA metabolism are altered in several metabolic disorders, including type 2 diabetes mellitus and cardiovascular diseases. In this review article, we present an overview of the current literature on the different effects of BCAAs in health and disease. We also highlight the results showing the most promising therapeutic effects of dietary BCAA supplementation and discuss how BCAAs can trigger different and even opposite effects, depending on the catabolic and anabolic states of the organisms. Moreover, we consider the effects of BCAAs when metabolism is abnormal, in the presence of a mixture of different anabolic and catabolic signals. These unique pharmacodynamic properties may partially explain some of the markedly different effects found in BCAA supplementation studies. To predict accurately these effects, the overall catabolic/anabolic status of patients should be carefully considered. In wider terms, a correct modulation of metabolic disorders would make nutraceutical interventions with BCAAs more effective. LINKED ARTICLES This article is part of a themed section on Principles of Pharmacological Research of Nutraceuticals. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.11/issuetoc.
Collapse
Affiliation(s)
- Francesco Bifari
- Laboratory of Cell Metabolism and Regenerative Medicine, Department of Medical Biotechnology and Translational MedicineUniversity of MilanMilanItaly
| | - Enzo Nisoli
- Center for Study and Research on Obesity, Department of Medical Biotechnology and Translational MedicineUniversity of MilanMilanItaly
| |
Collapse
|
27
|
Targeting endothelial metabolism for anti-angiogenesis therapy: A pharmacological perspective. Vascul Pharmacol 2017; 90:8-18. [DOI: 10.1016/j.vph.2017.01.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Accepted: 01/02/2017] [Indexed: 12/19/2022]
|
28
|
Soumeh EA, Hedemann MS, Poulsen HD, Corrent E, van Milgen J, Nørgaard JV. Nontargeted LC-MS Metabolomics Approach for Metabolic Profiling of Plasma and Urine from Pigs Fed Branched Chain Amino Acids for Maximum Growth Performance. J Proteome Res 2016; 15:4195-4207. [PMID: 27704848 DOI: 10.1021/acs.jproteome.6b00184] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The metabolic response in plasma and urine of pigs when feeding an optimum level of branched chain amino acids (BCAAs) for best growth performance is unknown. The objective of the current study was to identify the metabolic phenotype associated with the BCAAs intake level that could be linked to the animal growth performance. Three dose-response studies were carried out to collect blood and urine samples from pigs fed increasing levels of Ile, Val, or Leu followed by a nontargeted LC-MS approach to characterize the metabolic profile of biofluids when dietary BCAAs are optimum for animal growth. Results showed that concentrations of plasma hypoxanthine and tyrosine (Tyr) were higher while concentrations of glycocholic acid, tauroursodeoxycholic acid, and taurocholic acid were lower when the dietary Ile was optimum. Plasma 3-methyl-2-oxovaleric acid and creatine were lower when dietary Leu was optimum. The optimum dietary Leu resulted in increased urinary excretion of ascorbic acid and choline and relatively decreased excretion of 2-aminoadipic acid, acetyl-dl-valine, Ile, 2-methylbutyrylglycine, and Tyr. In conclusion, plasma glycocholic acid and taurocholic acid were discriminating metabolites to the optimum dietary Ile. The optimum dietary Leu was associated with reduced plasma creatine and urinary 2-aminoadipic acid and elevated urinary excretion of ascorbic acid and choline. The optimum dietary Val had a less pronounced metabolic response reflected in plasma or urine than other BCAA.
Collapse
Affiliation(s)
- Elham A Soumeh
- Department of Animal Science, Aarhus University, Foulum , DK-8830 Tjele, Denmark
| | - Mette S Hedemann
- Department of Animal Science, Aarhus University, Foulum , DK-8830 Tjele, Denmark
| | - Hanne D Poulsen
- Department of Animal Science, Aarhus University, Foulum , DK-8830 Tjele, Denmark
| | | | | | - Jan V Nørgaard
- Department of Animal Science, Aarhus University, Foulum , DK-8830 Tjele, Denmark
| |
Collapse
|
29
|
Yao K, Duan Y, Li F, Tan B, Hou Y, Wu G, Yin Y. Leucine in Obesity: Therapeutic Prospects. Trends Pharmacol Sci 2016; 37:714-727. [PMID: 27256112 DOI: 10.1016/j.tips.2016.05.004] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 05/08/2016] [Accepted: 05/10/2016] [Indexed: 02/07/2023]
|
30
|
Intracellular sources of ornithine for polyamine synthesis in endothelial cells. Amino Acids 2016; 48:2401-10. [DOI: 10.1007/s00726-016-2256-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2016] [Accepted: 05/06/2016] [Indexed: 01/26/2023]
|
31
|
McGarrity S, Halldórsson H, Palsson S, Johansson PI, Rolfsson Ó. Understanding the Causes and Implications of Endothelial Metabolic Variation in Cardiovascular Disease through Genome-Scale Metabolic Modeling. Front Cardiovasc Med 2016; 3:10. [PMID: 27148541 PMCID: PMC4834436 DOI: 10.3389/fcvm.2016.00010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 04/03/2016] [Indexed: 01/04/2023] Open
Abstract
High-throughput biochemical profiling has led to a requirement for advanced data interpretation techniques capable of integrating the analysis of gene, protein, and metabolic profiles to shed light on genotype-phenotype relationships. Herein, we consider the current state of knowledge of endothelial cell (EC) metabolism and its connections to cardiovascular disease (CVD) and explore the use of genome-scale metabolic models (GEMs) for integrating metabolic and genomic data. GEMs combine gene expression and metabolic data acting as frameworks for their analysis and, ultimately, afford mechanistic understanding of how genetic variation impacts metabolism. We demonstrate how GEMs can be used to investigate CVD-related genetic variation, drug resistance mechanisms, and novel metabolic pathways in ECs. The application of GEMs in personalized medicine is also highlighted. Particularly, we focus on the potential of GEMs to identify metabolic biomarkers of endothelial dysfunction and to discover methods of stratifying treatments for CVDs based on individual genetic markers. Recent advances in systems biology methodology, and how these methodologies can be applied to understand EC metabolism in both health and disease, are thus highlighted.
Collapse
Affiliation(s)
- Sarah McGarrity
- Center for Systems Biology, University of Iceland , Reykjavik , Iceland
| | - Haraldur Halldórsson
- Department of Pharmacology and Toxicology, School of Health Sciences, University of Iceland , Reykjavik , Iceland
| | - Sirus Palsson
- Center for Systems Biology, University of Iceland, Reykjavik, Iceland; Sinopia Biosciences Inc., San Diego, CA, USA
| | - Pär I Johansson
- Section for Transfusion Medicine, Capital Region Blood Bank, Rigshospitalet, University of Copenhagen , Copenhagen , Denmark
| | - Óttar Rolfsson
- Center for Systems Biology, University of Iceland, Reykjavik, Iceland; Department of Biochemistry and Molecular Biology, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| |
Collapse
|
32
|
Ruiz-Canela M, Toledo E, Clish CB, Hruby A, Liang L, Salas-Salvadó J, Razquin C, Corella D, Estruch R, Ros E, Fitó M, Gómez-Gracia E, Arós F, Fiol M, Lapetra J, Serra-Majem L, Martínez-González MA, Hu FB. Plasma Branched-Chain Amino Acids and Incident Cardiovascular Disease in the PREDIMED Trial. Clin Chem 2016; 62:582-592. [PMID: 26888892 PMCID: PMC4896732 DOI: 10.1373/clinchem.2015.251710] [Citation(s) in RCA: 218] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 01/08/2016] [Indexed: 12/13/2022]
Abstract
BACKGROUND The role of branched-chain amino acids (BCAAs) in cardiovascular disease (CVD) remains poorly understood. We hypothesized that baseline BCAA concentrations predict future risk of CVD and that a Mediterranean diet (MedDiet) intervention may counteract this effect. METHODS We developed a case-cohort study within the Prevención con Dieta Mediterránea (PREDIMED), with 226 incident CVD cases and 744 noncases. We used LC-MS/MS to measure plasma BCAAs (leucine, isoleucine, and valine), both at baseline and after 1 year of follow-up. The primary outcome was a composite of incident stroke, myocardial infarction, or cardiovascular death. RESULTS After adjustment for potential confounders, baseline leucine and isoleucine concentrations were associated with higher CVD risk: the hazard ratios (HRs) for the highest vs lowest quartile were 1.70 (95% CI, 1.05-2.76) and 2.09 (1.27-3.44), respectively. Stronger associations were found for stroke. For both CVD and stroke, we found higher HRs across successive quartiles of BCAAs in the control group than in the MedDiet groups. With stroke as the outcome, a significant interaction (P = 0.009) between baseline BCAA score and intervention with MedDiet was observed. No significant effect of the intervention on 1-year changes in BCAAs or any association between 1-year changes in BCAAs and CVD were observed. CONCLUSIONS Higher concentrations of baseline BCAAs were associated with increased risk of CVD, especially stroke, in a high cardiovascular risk population. A Mediterranean-style diet had a negligible effect on 1-year changes in BCAAs, but it may counteract the harmful effects of BCAAs on stroke.
Collapse
Affiliation(s)
- Miguel Ruiz-Canela
- Department of Preventive Medicine and Public Health, University of Navarra, Pamplona, Spain; IdiSNA, Navarra Institute for Health Research, Pamplona, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBER Obn), Instituto de Salud Carlos III, Madrid, Spain
| | - Estefania Toledo
- Department of Preventive Medicine and Public Health, University of Navarra, Pamplona, Spain; IdiSNA, Navarra Institute for Health Research, Pamplona, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBER Obn), Instituto de Salud Carlos III, Madrid, Spain
| | - Clary B Clish
- Broad Institute of MIT and Harvard University, Cambridge, MA
| | | | - Liming Liang
- Biostatistics, and Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Jordi Salas-Salvadó
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBER Obn), Instituto de Salud Carlos III, Madrid, Spain; Human Nutrition Department, IISPV, Universitat Rovira i Virgili, Reus, Spain
| | - Cristina Razquin
- Department of Preventive Medicine and Public Health, University of Navarra, Pamplona, Spain; IdiSNA, Navarra Institute for Health Research, Pamplona, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBER Obn), Instituto de Salud Carlos III, Madrid, Spain
| | - Dolores Corella
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBER Obn), Instituto de Salud Carlos III, Madrid, Spain; Department of Preventive Medicine, University of Valencia, Valencia, Spain
| | - Ramón Estruch
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBER Obn), Instituto de Salud Carlos III, Madrid, Spain; Department of Internal Medicine and
| | - Emilio Ros
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBER Obn), Instituto de Salud Carlos III, Madrid, Spain; Lipid Clinic, Department of Endocrinology and Nutrition Institut d'Investigacions Biomediques August Pi Sunyer (IDIBAPS), Hospital Clinic, University of Barcelona, Barcelona, Spain
| | - Montserrat Fitó
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBER Obn), Instituto de Salud Carlos III, Madrid, Spain; Cardiovascular and Nutrition Research Group, Institut de Recerca Hospital del Mar, Barcelona, Spain
| | - Enrique Gómez-Gracia
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBER Obn), Instituto de Salud Carlos III, Madrid, Spain; Department of Preventive Medicine, University of Malaga, Malaga, Spain
| | - Fernando Arós
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBER Obn), Instituto de Salud Carlos III, Madrid, Spain; Department of Cardiology, University Hospital of Alava, Vitoria, Spain
| | - Miquel Fiol
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBER Obn), Instituto de Salud Carlos III, Madrid, Spain; Institute of Health Sciences IUNICS, University of Balearic Islands and Hospital Son Espases, Palma de Mallorca, Spain
| | - José Lapetra
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBER Obn), Instituto de Salud Carlos III, Madrid, Spain; Department of Family Medicine, Primary Care Division of Sevilla, San Pablo Health Center, Sevilla, Spain
| | - Lluis Serra-Majem
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBER Obn), Instituto de Salud Carlos III, Madrid, Spain; Department of Clinical Sciences and Research Institute of Biomedical & Health Sciences, University of Las Palmas de Gran Canaria, Las Palmas, Spain
| | - Miguel A Martínez-González
- Department of Preventive Medicine and Public Health, University of Navarra, Pamplona, Spain; IdiSNA, Navarra Institute for Health Research, Pamplona, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBER Obn), Instituto de Salud Carlos III, Madrid, Spain
| | - Frank B Hu
- Departments of Nutrition, Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA; Channing Laboratory, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Cambridge, MA.
| |
Collapse
|
33
|
Solomon TM, Leech J, deBros GB, Murphy CA, Budson AE, Vassey EA, Solomon PR. A randomized, double-blind, placebo controlled, parallel group, efficacy study of alpha BRAIN® administered orally. Hum Psychopharmacol 2016; 31:135-43. [PMID: 26876224 DOI: 10.1002/hup.2520] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 12/16/2015] [Indexed: 11/11/2022]
Abstract
OBJECTIVE Alpha BRAIN® is a nootropic supplement that purports to enhance cognitive functioning in healthy adults. The goal of this study was to investigate the efficacy of this self-described cognitive enhancing nootropic on cognitive functioning in a group of healthy adults by utilizing a randomized, double blind, placebo-controlled design. METHODS A total of 63-treatment naïve individuals between 18 and 35 years of age completed the randomized, double-blind, placebo controlled trial. All participants completed a 2-week placebo run in before receiving active product, Alpha BRAIN® or new placebo, for 6 weeks. Participants undertook a battery of neuropsychological tests at randomization and at study completion. Primary outcome measures included a battery of neuropsychological tests and measures of sleep. RESULTS Compared with placebo, Alpha BRAIN® significantly improved on tasks of delayed verbal recall and executive functioning. Results also indicated significant time-by-group interaction in delayed verbal recall for the Alpha BRAIN® group. CONCLUSIONS The use of Alpha BRAIN® for 6 weeks significantly improved recent verbal memory when compared with controls, in a group of healthy adults. While the outcome of the study is encouraging, this is the first randomized controlled trial of Alpha BRAIN®, and the results merit further study.
Collapse
Affiliation(s)
- Todd M Solomon
- Boston Center for Memory, Newton, MA, USA.,The Memory Clinic, Bennington, VT, USA.,Boston University School of Medicine, Boston, MA, USA
| | | | | | - Cynthia A Murphy
- Boston Center for Memory, Newton, MA, USA.,The Memory Clinic, Bennington, VT, USA
| | - Andrew E Budson
- Boston Center for Memory, Newton, MA, USA.,Boston University School of Medicine, Boston, MA, USA
| | - Elizabeth A Vassey
- Boston Center for Memory, Newton, MA, USA.,Boston University School of Medicine, Boston, MA, USA
| | - Paul R Solomon
- Boston Center for Memory, Newton, MA, USA.,The Memory Clinic, Bennington, VT, USA.,Williams College, Williamstown, MA, USA
| |
Collapse
|
34
|
Alterations of amino acid metabolism in osteoarthritis: its implications for nutrition and health. Amino Acids 2016; 48:907-914. [DOI: 10.1007/s00726-015-2168-x] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 12/30/2015] [Indexed: 01/15/2023]
|
35
|
He X, Duan Y, Yao K, Li F, Hou Y, Wu G, Yin Y. β-Hydroxy-β-methylbutyrate, mitochondrial biogenesis, and skeletal muscle health. Amino Acids 2015; 48:653-664. [PMID: 26573541 DOI: 10.1007/s00726-015-2126-7] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Accepted: 11/02/2015] [Indexed: 12/16/2022]
Abstract
The metabolic roles of mitochondria go far beyond serving exclusively as the major producer of ATP in tissues and cells. Evidence has shown that mitochondria may function as a key regulator of skeletal muscle fiber types and overall well-being. Maintaining skeletal muscle mitochondrial content and function is important for sustaining health throughout the lifespan. Of great importance, β-hydroxy-β-methylbutyrate (HMB, a metabolite of L-leucine) has been proposed to enhance the protein deposition and efficiency of mitochondrial biogenesis in skeletal muscle, as well as muscle strength in both exercise and clinical settings. Specifically, dietary supplementation with HMB increases the gene expression of peroxisome proliferator-activated receptor gamma co-activator 1-alpha (PGC-1α), which represents an upstream inducer of genes of mitochondrial metabolism, coordinates the expression of both nuclear- and mitochondrion-encoded genes in mitochondrial biogenesis. Additionally, PGC-1α plays a key role in the transformation of skeletal muscle fiber type, leading to a shift toward type I muscle fibers that are rich in mitochondria and have a high capacity for oxidative metabolism. As a nitrogen-free metabolite, HMB holds great promise to improve skeletal muscle mass and function, as well as whole-body health and well-being of animals and humans.
Collapse
Affiliation(s)
- Xi He
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, 410128, China.,Hunan Co-Innovation Center of Animal Production Safety, Changsha, 410128, China
| | - Yehui Duan
- Scientific Observation and Experiment Station of Animal Nutrition and Feed Science in South-Central China, Ministry of Agriculture, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, No. 644 Yuanda Road, Furong District, Changsha, 410125, Hunan, China.,University of Chinese Academy of Sciences, Beijing, 100039, China
| | - Kang Yao
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, 410128, China. .,Scientific Observation and Experiment Station of Animal Nutrition and Feed Science in South-Central China, Ministry of Agriculture, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, No. 644 Yuanda Road, Furong District, Changsha, 410125, Hunan, China.
| | - Fengna Li
- Scientific Observation and Experiment Station of Animal Nutrition and Feed Science in South-Central China, Ministry of Agriculture, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, No. 644 Yuanda Road, Furong District, Changsha, 410125, Hunan, China
| | - Yongqing Hou
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, 430023, China
| | - Guoyao Wu
- Scientific Observation and Experiment Station of Animal Nutrition and Feed Science in South-Central China, Ministry of Agriculture, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, No. 644 Yuanda Road, Furong District, Changsha, 410125, Hunan, China.,Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, 430023, China.,Department of Animal Science, Texas A&M University, College Station, TX, 77843, USA
| | - Yulong Yin
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, 410128, China. .,Scientific Observation and Experiment Station of Animal Nutrition and Feed Science in South-Central China, Ministry of Agriculture, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, No. 644 Yuanda Road, Furong District, Changsha, 410125, Hunan, China. .,Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, 430023, China.
| |
Collapse
|
36
|
Ji Y, Wu Z, Dai Z, Sun K, Wang J, Wu G. Nutritional epigenetics with a focus on amino acids: implications for the development and treatment of metabolic syndrome. J Nutr Biochem 2015; 27:1-8. [PMID: 26427799 DOI: 10.1016/j.jnutbio.2015.08.003] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Revised: 07/31/2015] [Accepted: 08/05/2015] [Indexed: 12/31/2022]
Abstract
Recent findings from human and animal studies indicate that maternal undernutrition or overnutrition affects covalent modifications of the fetal genome and its associated histones that can be carried forward to subsequent generations. An adverse outcome of maternal malnutrition is the development of metabolic syndrome, which is defined as a cluster of disorders including obesity, hyperglycemia, hyperinsulinemia, hyperlipidemia, hypertension and insulin resistance. The transgenerational impacts of maternal nutrition are known as fetal programming, which is mediated by stable and heritable alterations of gene expression through covalent modifications of DNA and histones without changes in DNA sequences (namely, epigenetics). The underlying mechanisms include chromatin remodeling, DNA methylation (occurring at the 5'-position of cytosine residues within CpG dinucleotides), histone modifications (acetylation, methylation, phosphorylation, ubiquitination and sumoylation) and expression and activity of small noncoding RNAs. The enzymes catalyzing these reactions include S-adenosylmethionine-dependent DNA and protein methyltransferases, DNA demethylases, histone acetylase (lysine acetyltransferase), general control nonderepressible 5 (GCN5)-related N-acetyltransferase (a superfamily of acetyltransferase) and histone deacetylase. Amino acids (e.g., glycine, histidine, methionine and serine) and vitamins (B6, B12 and folate) play key roles in provision of methyl donors for DNA and protein methylation. Therefore, these nutrients and related metabolic pathways are of interest in dietary treatment of metabolic syndrome. Intervention strategies include targeting epigenetically disturbed metabolic pathways through dietary supplementation with nutrients (particularly functional amino acids and vitamins) to regulate one-carbon-unit metabolism, antioxidative reactions and gene expression, as well as protein methylation and acetylation. These mechanism-based approaches may effectively improve health and well-being of affected offspring.
Collapse
Affiliation(s)
- Yun Ji
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing 100193, China
| | - Zhenlong Wu
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing 100193, China.
| | - Zhaolai Dai
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing 100193, China
| | - Kaiji Sun
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing 100193, China
| | - Junjun Wang
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing 100193, China
| | - Guoyao Wu
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing 100193, China; Department of Animal Science and Center for Animal Genomics, Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
37
|
The role of leucine and its metabolites in protein and energy metabolism. Amino Acids 2015; 48:41-51. [DOI: 10.1007/s00726-015-2067-1] [Citation(s) in RCA: 186] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2015] [Accepted: 07/29/2015] [Indexed: 01/30/2023]
|
38
|
Safety of long-term dietary supplementation with L-arginine in rats. Amino Acids 2015; 47:1909-20. [PMID: 25948162 DOI: 10.1007/s00726-015-1992-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 04/13/2015] [Indexed: 02/05/2023]
Abstract
This study was conducted with rats to determine the safety of long-term dietary supplementation with L-arginine. Beginning at 6 weeks of age, male and female rats were fed a casein-based semi-purified diet containing 0.61 % L-arginine and received drinking water containing L-arginine-HCl (0, 1.8, or 3.6 g L-arginine/kg body-weight/day; n = 10/group). These supplemental doses of L-arginine were equivalent to 0, 286, and 573 mg L-arginine/kg body-weight/day, respectively, in humans. After a 13-week supplementation period, blood samples were obtained from rats for biochemical analyses. Supplementation with L-arginine increased plasma concentrations of arginine, ornithine, proline, homoarginine, urea, and nitric oxide metabolites without affecting those for lysine, histidine, or methylarginines, while reducing plasma concentrations of ammonia, glutamine, free fatty acids, and triglycerides. L-Arginine supplementation enhanced protein gain and reduced white-fat deposition in the body. Based on general appearance, feeding behavior, and physiological parameters, all animals showed good health during the entire experimental period; Plasma concentrations of all measured hormones (except leptin) did not differ between control and arginine-supplemented rats. L-Arginine supplementation reduced plasma levels of leptin. Additionally, L-arginine supplementation increased L-arginine:glycine amidinotransferase activity in kidneys but not in the liver or small intestine, suggesting tissue-specific regulation of enzyme expression by L-arginine. Collectively, these results indicate that dietary supplementation with L-arginine (e.g., 3.6 g/kg body-weight/day) is safe in rats for at least 91 days. This dose is equivalent to 40 g L-arginine/kg body-weight/day for a 70-kg person. Our findings help guide clinical studies to determine the safety of long-term oral administration of L-arginine to humans.
Collapse
|