1
|
Fan L, Chen J, Zhang Q, Ren J, Chen Y, Yang J, Wang L, Guo Z, Bu P, Zhu B, Zhao Y, Wang Y, Liu X, Wang W, Chen Z, Gao Q, Zheng L, Cai J. Fecal microbiota transplantation for hypertension: an exploratory, multicenter, randomized, blinded, placebo-controlled trial. MICROBIOME 2025; 13:133. [PMID: 40410854 PMCID: PMC12100813 DOI: 10.1186/s40168-025-02118-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 04/17/2025] [Indexed: 05/25/2025]
Abstract
BACKGROUND On the basis of the contribution of the gut microbiota to hypertension development, a novel strategy involving fecal microbiota transplantation (FMT) has been proposed to treat hypertension, but its efficacy has not been investigated in the clinic. METHODS In a randomized, blinded, placebo-controlled clinical trial (2021/03-2021/12, ClinicalTrials.gov, NCT04406129), hypertensive patients were recruited from seven centers in China, and received FMT or placebo capsules orally at three visits. The patients were randomized at a 1:1 ratio in blocks of four and stratified by center by an independent statistician. The intention-to-treat principle was implemented, as all randomized participants who received at least one intervention were included. The primary outcome was the decrease in office systolic blood pressure (SBP) from baseline to the day 30 visit. Adverse events (AEs) were recorded through the 3-month follow-up to assess safety measures. Alterations in BP, the fecal microbiome, and the plasma metabolome were assessed via exploratory analyses. RESULTS This study included 124 patients (mean age 43 years, 73.4% men) who received FMT (n = 63) or placebo (n = 61) capsules. The numbers of participants who experienced AEs (13 (20.6%) vs. 9 (14.8%), p = 0.39) and the primary outcome (6.28 (11.83) vs. 5.77 (10.06) mmHg, p = 0.62) were comparable between the groups. The FMT group presented a decrease in SBP after 1 week of FMT, with a between-arm difference of - 4.34 (95% CI, - 8.1 to - 0.58; p = 0.024) mmHg, but this difference did not persist even after repeated intervention. After FMT, shifts in microbial richness and structure were identified and the abundance of the phyla Firmicutes and Bacteroidetes was altered. Decreases in the abundances of Eggerthella lenta, Erysipelatoclostridium ramosum, Anaerostipes hadrus, Gemella haemolysans, and Streptococcus vestibularis and increases in the abundances of Parabacteroides merdae, Prevotella copri, Bacteroides galacturonicus, Eubacterium sp. CAG 180, Desulfovibrio piger, Megamonas hypermegale, Collinsella stercoris, Coprococcus catus, and Allisonella histaminiformans were identified and correlated with office SBP. Those species were also correlated with responding and inversely office SBP-associated metabolites including tyrosine, glutamine, aspartate, phenylalanine, methionine, serine, sarcosine, and/or asparagine. CONCLUSIONS Safety but unsustainable BP reduction was observed in the first trial of the effects of FMT on hypertension. Additional intervention studies on specific microbes with metabolite-targeting and BP-modulating features are needed. Video Abstract.
Collapse
Affiliation(s)
- Luyun Fan
- Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Chinese Institutes for Medical Research, Beijing, 100029, China
| | - Junru Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao, China
- Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, 410078, China
| | - Qi Zhang
- The Institute of Cardiovascular Sciences and Institute of Systems Biomedicine, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Health Science Center, Peking University, Beijing, 100191, China
| | - Jie Ren
- Shanxi Bethune Hospital, Taiyuan, 030032, Shanxi, China
| | - Youren Chen
- Department of Cardiology, Second Affiliated Hospital of Shantou University Medical College, Shantou, 515000, Guangdong, China
| | - Jinfeng Yang
- The People's Hospital of Ji Xian District, Tianjin, 301900, China
| | - Lu Wang
- Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Chinese Institutes for Medical Research, Beijing, 100029, China
| | - Zihong Guo
- Fuwai Yunnan Cardiovascular Hospital, Kunming, Yunnan, China
| | - Peili Bu
- Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Bingpo Zhu
- Southern University of Science and Technology Hospital, Shenzhen, China
| | - Yanyan Zhao
- Medical Research & Biometrics Center, National Center for Cardiovascular Dieases, Fuwai Hospital Chinese Academy of Medical Sciences, Beijing, 100037, China
| | - Yang Wang
- Medical Research & Biometrics Center, National Center for Cardiovascular Dieases, Fuwai Hospital Chinese Academy of Medical Sciences, Beijing, 100037, China
| | - Xiaoyan Liu
- Department of Cardiology, Heart Center, Beijing, Key Laboratory of Hypertension Research, Medical Research Center, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Wenjie Wang
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Zhenzhen Chen
- Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Chinese Institutes for Medical Research, Beijing, 100029, China
| | - Qiannan Gao
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Lemin Zheng
- Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, 410078, China.
| | - Jun Cai
- Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Chinese Institutes for Medical Research, Beijing, 100029, China.
| |
Collapse
|
2
|
Sousa A, Oliveira N, Conde R, Morais E, Amaral AP, Embade N, Millet O, Verde I. Nuclear Magnetic Resonance Analysis Seeking for Metabolic Markers of Hypertension in Human Serum. Molecules 2025; 30:2145. [PMID: 40430317 PMCID: PMC12113710 DOI: 10.3390/molecules30102145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2025] [Revised: 05/08/2025] [Accepted: 05/10/2025] [Indexed: 05/29/2025] Open
Abstract
Hypertension is a highly prevalent medical condition that occurs when blood pressure is too high, which greatly increases the risk of developing other cardiovascular diseases and is generally associated with higher rates of morbidity and mortality. Due to the silent/asymptomatic nature of hypertension, although the methods currently available to diagnose it are easy, they generally do not allow for an early diagnosis and an efficient prognosis to avoid irreversible damage in the medium or long term. In fact, an early diagnosis of hypertension would be crucial to decrease hypertension-associated mortality. Since metabolomics using NMR can provide a global measurement of various serum metabolites, it is very suitable for detecting novel biomarkers. We therefore analyzed serum metabolomic profiles among normotensive and hypertensive elderly individuals by NMR and identified new potential biomarkers for hypertension and associated diseases. We found higher levels of acetate, formate, and glycerol, and lower levels of glutamine, glycine, and sarcosine in individuals with hypertension. Therefore, these metabolites could be used for early diagnosis of hypertension to avoid comorbidities derived from hypertension and associated mortality.
Collapse
Affiliation(s)
- Adriana Sousa
- Health Sciences Research Centre (CICS-UBI), University of Beira Interior (UBI), 6200-506 Covilhã, Portugal; (A.S.); (N.O.); (R.C.); (E.M.); (A.P.A.)
- RISE-Health, Faculty of Health Sciences, University of Beira Interior (UBI), Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Nádia Oliveira
- Health Sciences Research Centre (CICS-UBI), University of Beira Interior (UBI), 6200-506 Covilhã, Portugal; (A.S.); (N.O.); (R.C.); (E.M.); (A.P.A.)
- RISE-Health, Faculty of Health Sciences, University of Beira Interior (UBI), Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Ricardo Conde
- Health Sciences Research Centre (CICS-UBI), University of Beira Interior (UBI), 6200-506 Covilhã, Portugal; (A.S.); (N.O.); (R.C.); (E.M.); (A.P.A.)
- Center for Cooperative Research in Biosciences (CIC-BioGUNE), Bizkaia Science and Technology Park, 48160 Derio, Spain; (N.E.); (O.M.)
| | - Elisabete Morais
- Health Sciences Research Centre (CICS-UBI), University of Beira Interior (UBI), 6200-506 Covilhã, Portugal; (A.S.); (N.O.); (R.C.); (E.M.); (A.P.A.)
| | - Ana Paula Amaral
- Health Sciences Research Centre (CICS-UBI), University of Beira Interior (UBI), 6200-506 Covilhã, Portugal; (A.S.); (N.O.); (R.C.); (E.M.); (A.P.A.)
| | - Nieves Embade
- Center for Cooperative Research in Biosciences (CIC-BioGUNE), Bizkaia Science and Technology Park, 48160 Derio, Spain; (N.E.); (O.M.)
| | - Oscar Millet
- Center for Cooperative Research in Biosciences (CIC-BioGUNE), Bizkaia Science and Technology Park, 48160 Derio, Spain; (N.E.); (O.M.)
| | - Ignacio Verde
- Health Sciences Research Centre (CICS-UBI), University of Beira Interior (UBI), 6200-506 Covilhã, Portugal; (A.S.); (N.O.); (R.C.); (E.M.); (A.P.A.)
- RISE-Health, Faculty of Health Sciences, University of Beira Interior (UBI), Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| |
Collapse
|
3
|
Zhao Q, Li J, Diao Z, Zhang X, Feng S, Hou G, Xu W, Zhao Z, Qiu Z, Yang W, Zhou S, Tian P, Zhang Q, Chen W, Li H, Xiao G, Qin J, Hu L, Li Z, Lin L, Wang S, Gao R, Huang W, Ruan X, Zhang S, Zhang J, Zhao L, Zhang R. Early prediction of preeclampsia from clinical, multi-omics and laboratory data using random forest model. BMC Pregnancy Childbirth 2025; 25:531. [PMID: 40325391 PMCID: PMC12051331 DOI: 10.1186/s12884-025-07582-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 04/08/2025] [Indexed: 05/07/2025] Open
Abstract
BACKGROUND Predicting preeclampsia (PE) within the first 16 weeks of gestation is difficult due to various risk factors, poorly understood causes and likely multiple pathogenic phenotypes of preeclampsia. OBJECTIVES: In this study, we aimed to develop prediction models for early-onset preeclampsia (EPE) and late-onset preeclampsia (LPE) respectively using clinical data, metabolome and proteome analyses on plasma samples and laboratory data. METHODS We retrospectively recruited 56 EPE, 50 LPE patients and 92 normotensive controls from three tertiary hospitals and used clinical and laboratory data in early pregnancy. Models for EPE and LPE were fitted with the use of patient' clinical, multi-omics and laboratory data. RESULTS By comparing multi-omics and laboratory test variables between EPE, LPE and healthy controls, we identified sets of differentially expressed biomarkers, including 49 and 33 metabolites, 28 and 36 proteins as well as 5 and 7 laboratory variables associated with EPE and LPE respectively. Using the random forest algorithm, we developed a prediction model using seven clinical factors, seven metabolites, five laboratory test variables. The model yielded the highest accuracy for EPE prediction with good sensitivity (87.5%, 95% confidence interval [CI]: 67.64%-97.34%) and specificity (94.1%, 95% CI: 80.32%-99.28%). We also developed a prediction model that exhibited high accuracy in separating LPE from controls (sensitivity: 66.67%, 95% CI: 43.03%-85.41%; specificity: 94.12%, 95% CI: 80.32%-99.28%) using seven clinical factors, five metabolites and eight proteins. CONCLUSION Our study has identified a set of significant omics and laboratory features for PE prediction. The established models yielded high prediction performance for preeclampsia risk from clinical, multi-omics and laboratory information.
Collapse
Affiliation(s)
- Qiang Zhao
- Department of Obstetrics and Gynecology, Jiangmen Central Hospital, Guangdong Province, Jiangmen, 529030, China
- Clinical Transformation and Application Key Lab for Obstetrics and Gynecology, Pediatrics, and Reproductive Medicine of Jiangmen, Guangdong Province, Jiangmen, 529030, China
| | - Jia Li
- Clin Lab, Hebei Industrial Technology Research Institute of Genomics in Maternal & Child Health, BGI Genomics, Hebei Province, Shijiazhuang, 050011, China
- BGI Genomics, Guangdong Province, Shenzhen, 518083, China
| | - Zhuo Diao
- BGI Genomics, Guangdong Province, Shenzhen, 518083, China
| | - Xiao Zhang
- Clin Lab, Hebei Industrial Technology Research Institute of Genomics in Maternal & Child Health, BGI Genomics, Hebei Province, Shijiazhuang, 050011, China
- BGI Genomics, Guangdong Province, Shenzhen, 518083, China
| | - Suihua Feng
- Department of Obstetrics and Gynecology, Jiangmen Central Hospital, Guangdong Province, Jiangmen, 529030, China
| | - Guixue Hou
- BGI Genomics, Guangdong Province, Shenzhen, 518083, China
| | - Wenqiu Xu
- Clin Lab, Hebei Industrial Technology Research Institute of Genomics in Maternal & Child Health, BGI Genomics, Hebei Province, Shijiazhuang, 050011, China
- BGI Genomics, Guangdong Province, Shenzhen, 518083, China
| | - Zhiguang Zhao
- Clin Lab, Hebei Industrial Technology Research Institute of Genomics in Maternal & Child Health, BGI Genomics, Hebei Province, Shijiazhuang, 050011, China
- BGI Genomics, Guangdong Province, Shenzhen, 518083, China
| | - Zhixu Qiu
- Clin Lab, Hebei Industrial Technology Research Institute of Genomics in Maternal & Child Health, BGI Genomics, Hebei Province, Shijiazhuang, 050011, China
- BGI Genomics, Guangdong Province, Shenzhen, 518083, China
| | - Wenzhi Yang
- Clin Lab, Hebei Industrial Technology Research Institute of Genomics in Maternal & Child Health, BGI Genomics, Hebei Province, Shijiazhuang, 050011, China
- BGI Genomics, Guangdong Province, Shenzhen, 518083, China
| | - Si Zhou
- Clin Lab, Hebei Industrial Technology Research Institute of Genomics in Maternal & Child Health, BGI Genomics, Hebei Province, Shijiazhuang, 050011, China
- BGI Genomics, Guangdong Province, Shenzhen, 518083, China
| | - Peirun Tian
- BGI Genomics, Guangdong Province, Shenzhen, 518083, China
| | - Qun Zhang
- Department of Obstetrics and Gynecology, Jiangmen Central Hospital, Guangdong Province, Jiangmen, 529030, China
| | - Weiping Chen
- Department of Obstetrics and Gynecology, Jiangmen Central Hospital, Guangdong Province, Jiangmen, 529030, China
| | - Huahua Li
- Department of Obstetrics and Gynecology, Jiangmen Central Hospital, Guangdong Province, Jiangmen, 529030, China
| | - Gefei Xiao
- Institute of Medical Genetics, Zhuhai Center for Maternal and Child Health Care, Guangdong Province, Zhuhai, 519000, China
| | - Jie Qin
- Institute of Medical Genetics, Zhuhai Center for Maternal and Child Health Care, Guangdong Province, Zhuhai, 519000, China
| | - Liqing Hu
- Institute of Medical Genetics, Zhuhai Center for Maternal and Child Health Care, Guangdong Province, Zhuhai, 519000, China
| | - Zhongzhe Li
- Department of Prevention and Health Care, Zhuhai Center for Maternal and Child Health Care, Guangdong Province, Zhuhai, 519000, China
| | - Liang Lin
- BGI Genomics, Guangdong Province, Shenzhen, 518083, China
| | - Shunyao Wang
- BGI Genomics, Guangdong Province, Shenzhen, 518083, China
- Hunan Provincial Key Laboratory of Regional Hereditary Birth Defects Prevention and Control, Changsha Hospital for Maternal & Child Health CareAffiliated to, Hunan Normal University , Hunan Province, Changsha, 410007, China
| | - Ruyun Gao
- School of Public Health, Hebei Province Key Laboratory of Environment and Human Health, Hebei Medical University, Hebei Province, Shijiazhuang, 050017, China
| | - Wuyan Huang
- Institute of Medical Genetics, Zhuhai Center for Maternal and Child Health Care, Guangdong Province, Zhuhai, 519000, China
| | - Xiaohong Ruan
- Department of Obstetrics and Gynecology, Jiangmen Central Hospital, Guangdong Province, Jiangmen, 529030, China.
- Clinical Transformation and Application Key Lab for Obstetrics and Gynecology, Pediatrics, and Reproductive Medicine of Jiangmen, Guangdong Province, Jiangmen, 529030, China.
| | - Sufen Zhang
- Institute of Medical Genetics, Zhuhai Center for Maternal and Child Health Care, Guangdong Province, Zhuhai, 519000, China.
| | - Jianguo Zhang
- Clin Lab, Hebei Industrial Technology Research Institute of Genomics in Maternal & Child Health, BGI Genomics, Hebei Province, Shijiazhuang, 050011, China.
- BGI Genomics, Guangdong Province, Shenzhen, 518083, China.
- School of Public Health, Hebei Medical University, Shijiazhuang, 050017, China.
| | - Lijian Zhao
- Clin Lab, Hebei Industrial Technology Research Institute of Genomics in Maternal & Child Health, BGI Genomics, Hebei Province, Shijiazhuang, 050011, China.
- BGI Genomics, Guangdong Province, Shenzhen, 518083, China.
- Medical Technology College, Hebei Medical University, Shijiazhuang, 050017, China.
| | - Rui Zhang
- Division of Maternal-Fetal Medicine, Shenzhen Baoan Women's and Children's Hospital, Guangdong Province, Shenzhen, 518102, China.
| |
Collapse
|
4
|
Tong TY, Li Y, Rexrode KM, Willett WC, Sun Q, Manson JE, Longo VD, Key TJ, Hu FB. Dietary Amino Acids and Risk of Stroke Subtypes: Results from 3 Large Prospective Cohort Studies. J Nutr 2025; 155:1560-1569. [PMID: 40127732 PMCID: PMC12121403 DOI: 10.1016/j.tjnut.2025.03.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 03/07/2025] [Accepted: 03/19/2025] [Indexed: 03/26/2025] Open
Abstract
BACKGROUND Differences in dietary protein have been associated with stroke risk, with possible heterogeneity in associations by stroke type or food sources of protein. OBJECTIVES We examined the associations of individual dietary amino acids, as the constituents of dietary protein, with risks of ischemic, hemorrhagic, and total stroke. METHODS We analyzed data from 73,830 females in the Nurses' Health Study (1984-2012), 92,333 females in the Nurses' Health Study II (1991-2013), and 43,268 males in the Health Professionals Follow-Up Study (1986-2016). Dietary intakes of 22 (20 standard and 2 nonstandard) amino acids were assessed using validated food frequency questionnaires, administered typically every 4 y. Multivariable-adjusted Cox regression models were used to estimate hazard ratios (HRs) and 95% confidence intervals (CIs) of ischemic, hemorrhagic, and total stroke in relation to the energy-adjusted intakes of individual amino acids. RESULTS During a mean follow-up of 23.7 y, 3058 ischemic, 872 hemorrhagic, and 5997 total stroke cases were documented. After correction for multiple testing, lower risks of ischemic stroke were observed with higher intakes of glutamine (HR per 1 standard deviation higher: 0.94, 95% CI: 0.90, 0.98, P = 0.004) and proline (0.94, 0.90, 0.98, P = 0.005). The associations remained directionally consistent across sensitivity analyses but attenuated upon mutual adjustment. All other amino acids, including branched-chain amino acids, were not significantly associated with ischemic stroke. For hemorrhagic stroke, no significant associations were observed for any of the amino acids. For total stroke, inverse associations were also observed for both glutamine (0.94, 0.91-0.97, P < 0.001) and proline (0.96, 0.93-0.99, P = 0.004). In terms of dietary sources, glutamine was most strongly correlated with plant protein and whole grains, whereas proline was most strongly correlated with dairy protein and dairy products. CONCLUSIONS Higher intakes of glutamine and proline were associated with lower risks of ischemic and total stroke.
Collapse
Affiliation(s)
- Tammy Yn Tong
- Cancer Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Yanping Li
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Kathryn M Rexrode
- Division of Women's Health, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Walter C Willett
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, United States; Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Qi Sun
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, United States; Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - JoAnn E Manson
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, United States; Division of Preventive Medicine Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Valter D Longo
- Longevity Institute, Davis School of Gerontology and Department of Biological Sciences, University of Southern California, Los Angeles, CA, United States; IFOM ETS, AIRC Institute of Molecular Oncology, Milan, Italy
| | - Timothy J Key
- Cancer Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Frank B Hu
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, United States; Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, United States.
| |
Collapse
|
5
|
Knauss HM, Ambatipudi M, Kosyakovsky LB, Herzig MS, Wang JK, Liu EE, Lau ES, McNeill JN, Shi X, Gerszten RE, Hamburg NM, Lewis GD, Robbins JM, Ho JE. Trans-right ventricle metabolite gradients in obesity highlight multiple metabolic pathways. Physiol Rep 2025; 13:e70323. [PMID: 40350961 PMCID: PMC12066817 DOI: 10.14814/phy2.70323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 01/30/2025] [Accepted: 02/19/2025] [Indexed: 05/14/2025] Open
Abstract
Obesity and metabolic dysfunction are associated with pulmonary vascular remodeling, yet molecular mechanisms remain poorly understood. We sought to study trans-right ventricular (RV) metabolite gradients to elucidate potential molecular pathways operant among individuals with obesity and pulmonary hypertension. In this study, 38 individuals with obesity (mean age 58 years, 68% women, average BMI 36.6 kg/m2) underwent invasive right heart catheterization. Multi-site blood sampling from the superior vena cava and pulmonary artery was performed to assess trans-RV gradients, with targeted metabolite profiling using liquid chromatography-mass spectrometry. We found 56 metabolites with significant trans-RV gradients (FDR q < 0.05), including intermediates of fatty acid oxidation, the tricarboxylic acid cycle, and nucleotide metabolism. Further, trans-RV gradients in lipid and purine metabolism were associated with BMI and related cardiometabolic traits, such as waist circumference, insulin resistance, and serum lipids. Finally, differential levels of bile acids, intermediates of lipid peroxidation, and nucleotide metabolism across the RV were associated with pulmonary hypertension. In conclusion, trans-RV metabolite gradients among individuals with obesity reveal alterations in metabolites representative of molecular pathways such as fatty acid oxidation, and others correlated with cardiometabolic traits and/or pulmonary hypertension, including orotic acid, bile acids, and acylcarnitines.
Collapse
Affiliation(s)
- Hanna M. Knauss
- Division of CardiologyBeth Israel Deaconess Medical CenterBostonMassachusettsUSA
| | - Mythri Ambatipudi
- Division of CardiologyBeth Israel Deaconess Medical CenterBostonMassachusettsUSA
| | - Leah B. Kosyakovsky
- Division of CardiologyBeth Israel Deaconess Medical CenterBostonMassachusettsUSA
| | - Matthew S. Herzig
- Division of CardiologyBeth Israel Deaconess Medical CenterBostonMassachusettsUSA
| | - Jessica K. Wang
- Division of CardiologyBeth Israel Deaconess Medical CenterBostonMassachusettsUSA
| | - Elizabeth E. Liu
- Division of CardiologyBeth Israel Deaconess Medical CenterBostonMassachusettsUSA
| | - Emily S. Lau
- Division of CardiologyMassachusetts General HospitalBostonMassachusettsUSA
| | - Jenna N. McNeill
- Division of Pulmonary and Critical Care MedicineDuke University HospitalDurhamNorth CarolinaUSA
| | - Xu Shi
- Division of CardiologyBeth Israel Deaconess Medical CenterBostonMassachusettsUSA
| | - Robert E. Gerszten
- Division of CardiologyBeth Israel Deaconess Medical CenterBostonMassachusettsUSA
| | - Naomi M. Hamburg
- Whitaker Cardiovascular Institute, School of MedicineBoston UniversityBostonMassachusettsUSA
| | - Gregory D. Lewis
- Division of CardiologyMassachusetts General HospitalBostonMassachusettsUSA
| | - Jeremy M. Robbins
- Division of CardiologyBeth Israel Deaconess Medical CenterBostonMassachusettsUSA
| | - Jennifer E. Ho
- Division of CardiologyBeth Israel Deaconess Medical CenterBostonMassachusettsUSA
| |
Collapse
|
6
|
Passadore MD, Azinheira Nobrega Cruz N, Bocato MZ, Ferreira LDA, Icimoto MY, Molina MDCB, Mill JG, Barbosa Junior F, Casarini DE, de Oliveira LCG. Urinary amino acid metabolomic profiling and its association with childhood obesity in prepubescent individuals. Front Physiol 2025; 16:1524939. [PMID: 40365082 PMCID: PMC12069889 DOI: 10.3389/fphys.2025.1524939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 04/07/2025] [Indexed: 05/15/2025] Open
Abstract
Introduction Amino acids are fundamental in several metabolic processes, and their levels can reflect metabolism impairments that contribute to obesity and related diseases. Our objective was to identify a urinary amino acid fingerprint in obese and overweight children in prepuberty and to correlate this profile with cardiometabolic alterations. Methods The study included 110 children, boys and girls aged 9-10 years, they were classified according to their BMI-for-age (Body Mass Index for age) into three groups: normal weight (NW) (n = 45), overweight (OW) (n = 21), and obese (OB) (n = 44). The 12-h urine samples were analyzed by LC-MS/MS to quantify 47 amino acids using the Amino Acids Analysis Kit (Zivak®, Turkey), values were corrected by creatinine concentration. Anthropometric measurements, cardiovascular parameters, and biochemical profiles were assessed following standard protocols. Results When compared to NW, anthropometric measures, systolic and diastolic blood pressure, and serum uric acid levels were progressively elevated in the OW and OB groups. The OB group was characterized by elevated alpha-aminoadipic acid, asparagine, cystathionine, 1-methyl-histidine, serine, tryptophan, phenylalanine, and tyrosine. In contrast, the OW group presented the most expressive levels of glutamine, alpha-diaminopimelic, and sarcosine. Discussion Our findings indicate that obese and overweight children exhibit a particular urinary amino acid fingerprint which is similar to that reported in studies with plasma. The altered amino acids, particularly tyrosine, are frequently associated with impairments in glucose homeostasis, insulin resistance, and diabetes mellitus type 2. Potential mechanisms for increasing the levels of these amino acids in excess of weight may include enhanced protein degradation and impaired oxidative metabolism.
Collapse
Affiliation(s)
- Mariana Doce Passadore
- Postgraduation Program of Nephrology, Nephrology Division, Department of Medicine, Universidade Federal de São Paulo (UNIFESP/EPM), Sao Paulo, Brazil
| | - Nayara Azinheira Nobrega Cruz
- Postgraduation Program of Translational Medicine, Nephrology Division, Department of Medicine, Universidade Federal de São Paulo (UNIFESP/EPM), Sao Paulo, Brazil
| | - Mariana Zuccherato Bocato
- Analytical and System Toxicology Laboratory, Department of Clinical, Toxicological and Bromatological Analysis, Faculty of Pharmaceutical Sciences of Ribeirao Preto, Universidade de São Paulo, Sao Paulo, Brazil
| | | | - Marcelo Yudi Icimoto
- Department of Biophysics, Universidade Federal de São Paulo (UNIFESP/EPM), Sao Paulo, Brazil
| | - Maria del Carmen Bisi Molina
- Department of Integrated Health Education, Center for Health Sciences, Universidade Federal do Espírito Santo, Vitória, Brazil
| | - José Geraldo Mill
- Department of Physiological Sciences, Universidade Federal do Espírito Santo, Vitória, Brazil
| | - Fernando Barbosa Junior
- Analytical and System Toxicology Laboratory, Department of Clinical, Toxicological and Bromatological Analysis, Faculty of Pharmaceutical Sciences of Ribeirao Preto, Universidade de São Paulo, Sao Paulo, Brazil
| | - Dulce Elena Casarini
- Nephrology Division, Department of Medicine, Universidade Federal de São Paulo (UNIFESP/EPM), Sao Paulo, Brazil
| | | |
Collapse
|
7
|
Makassy D, Williams K, Karwi QG. The Evolving Role of Macrophage Metabolic Reprogramming in Obesity. Can J Cardiol 2025:S0828-282X(25)00320-4. [PMID: 40311669 DOI: 10.1016/j.cjca.2025.04.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 04/17/2025] [Accepted: 04/21/2025] [Indexed: 05/03/2025] Open
Abstract
Recent research has extensively explored the critical role of energy metabolism in shaping the inflammatory response and polarization of macrophages in obesity. This rapidly growing field emphasizes the need to understand the connection between metabolic processes that support macrophage polarization in obesity. Although most published research in this area has focused on glucose and fatty acids, how the flux through other metabolic pathways (such as ketone and amino acid oxidation) in macrophages is altered in obesity is not well defined. This review summarizes the main alterations in uptake, storage, and oxidation of oxidative substrates (glucose, fatty acids, ketone bodies, and amino acids) in macrophages and how these alterations are linked to macrophage polarization and contribution to augmented inflammatory markers in obesity. The review also discusses how oxidative substrates could modulate macrophage energy metabolism and inflammatory responses via feeding into other nonoxidative pathways (such as the pentose phosphate pathway, triacylglycerol synthesis/accumulation), via acting as signalling molecules, or via mediating post-translational modifications (such as O-GlcNAcylation or β-hydroxybutyrylation). The review also identifies several critical unanswered questions regarding the characteristics (functional and metabolic) of macrophages from different origins (adipose tissue, skeletal muscle, bone marrow) in obesity and how these characteristics contribute to early vs late phases of obesity. We also identified a number of new therapeutic targets that could be evaluated in future investigations. Targeting macrophage metabolism in obesity is an exciting and active area of research with significant potential to help identify new treatments to limit the detrimental effects of inflammation in obesity.
Collapse
Affiliation(s)
- Dorcus Makassy
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, Saint John's, Newfoundland and Labrador, Canada
| | - Kyra Williams
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, Saint John's, Newfoundland and Labrador, Canada
| | - Qutuba G Karwi
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, Saint John's, Newfoundland and Labrador, Canada.
| |
Collapse
|
8
|
Yang X, Zhang F, Zhang B, Qi H, Xie Y, Peng W, Li B, Wen F, Li P, Sun Y, Qu A, Zhang L. Associations of Metabolites Related Salt Sensitivity of Blood Pressure and Essential Hypertension in Chinese Population: The EpiSS Study. Nutrients 2025; 17:1289. [PMID: 40219046 PMCID: PMC11990569 DOI: 10.3390/nu17071289] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Revised: 04/03/2025] [Accepted: 04/03/2025] [Indexed: 04/14/2025] Open
Abstract
BACKGROUND Salt sensitivity of blood pressure (SSBP) is an important risk factor for essential hypertension and cardiovascular diseases, and its metabolic mechanisms remain poorly understood. This study aimed to identify SSBP-associated metabolic biomarkers and investigate their potential mediating role in the SSBP-hypertension pathophysiology. METHODS Based on the Systematic Epidemiological Study of Salt Sensitivity (EpiSS) conducted in 2014-2016, we performed a case-control study involving 54 matched pairs of participants classified as salt-sensitive or salt-resistant with targeted metabolomics detected. Multivariable logistic regression analyses were conducted to assess the metabolites associations with SSBP and hypertension. The diagnostic performance of the model was evaluated using the receiver operating characteristic curve (ROC) analysis yielded an area under the curve (AUC) value, sensitivity, and specificity. Furthermore, the potential mediating effects of targeted metabolites on the relationship between SSBP and essential hypertension were explored. RESULTS Three metabolites demonstrated significant SSBP associations: L-Glutamine (OR = 0.998; 95% CI: 0.997, 0.999), PC (16:1/14:0) (OR = 1.039; 95% CI: 1.003, 1.077), and ChE (22:4) (OR = 1.115; 95% CI: 1.002, 1.240). Among them, L-Glutamine demonstrated the highest diagnostic efficiency for SSBP (AUC = 0.766; 95% CI: 0.677, 0.855). The combined model of the three metabolites slightly improved diagnostic efficiency (AUC = 0.788; 95% CI: 0.703, 0.874). L-Glutamine and Cer (d18:0/24:1) were identified as potential protective factors against essential hypertension (p < 0.05). Mediation analyses further indicated that L-Glutamine partially mediated the relationship between SSBP and essential hypertension, demonstrating a suppressive effect. CONCLUSIONS This study identified L-Glutamine as both a diagnostic biomarker for SSBP and a metabolic modulator attenuating hypertension risk, providing insights for early SSBP screening and the pathways governing SSBP progression to overt hypertension.
Collapse
Affiliation(s)
- Xiaojun Yang
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, and Beijing Key Laboratory of Environment and Aging, Beijing 100069, China; (X.Y.); (F.Z.); (B.Z.); (H.Q.); (Y.X.); (W.P.); (B.L.); (F.W.); (P.L.); (Y.S.); (A.Q.)
| | - Fengxu Zhang
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, and Beijing Key Laboratory of Environment and Aging, Beijing 100069, China; (X.Y.); (F.Z.); (B.Z.); (H.Q.); (Y.X.); (W.P.); (B.L.); (F.W.); (P.L.); (Y.S.); (A.Q.)
- Health Management Center, Beijing Aerospace General Hospital, Beijing 100076, China
| | - Bowen Zhang
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, and Beijing Key Laboratory of Environment and Aging, Beijing 100069, China; (X.Y.); (F.Z.); (B.Z.); (H.Q.); (Y.X.); (W.P.); (B.L.); (F.W.); (P.L.); (Y.S.); (A.Q.)
| | - Han Qi
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, and Beijing Key Laboratory of Environment and Aging, Beijing 100069, China; (X.Y.); (F.Z.); (B.Z.); (H.Q.); (Y.X.); (W.P.); (B.L.); (F.W.); (P.L.); (Y.S.); (A.Q.)
| | - Yunyi Xie
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, and Beijing Key Laboratory of Environment and Aging, Beijing 100069, China; (X.Y.); (F.Z.); (B.Z.); (H.Q.); (Y.X.); (W.P.); (B.L.); (F.W.); (P.L.); (Y.S.); (A.Q.)
| | - Wenjuan Peng
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, and Beijing Key Laboratory of Environment and Aging, Beijing 100069, China; (X.Y.); (F.Z.); (B.Z.); (H.Q.); (Y.X.); (W.P.); (B.L.); (F.W.); (P.L.); (Y.S.); (A.Q.)
| | - Bingxiao Li
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, and Beijing Key Laboratory of Environment and Aging, Beijing 100069, China; (X.Y.); (F.Z.); (B.Z.); (H.Q.); (Y.X.); (W.P.); (B.L.); (F.W.); (P.L.); (Y.S.); (A.Q.)
| | - Fuyuan Wen
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, and Beijing Key Laboratory of Environment and Aging, Beijing 100069, China; (X.Y.); (F.Z.); (B.Z.); (H.Q.); (Y.X.); (W.P.); (B.L.); (F.W.); (P.L.); (Y.S.); (A.Q.)
| | - Pandi Li
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, and Beijing Key Laboratory of Environment and Aging, Beijing 100069, China; (X.Y.); (F.Z.); (B.Z.); (H.Q.); (Y.X.); (W.P.); (B.L.); (F.W.); (P.L.); (Y.S.); (A.Q.)
| | - Yuan Sun
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, and Beijing Key Laboratory of Environment and Aging, Beijing 100069, China; (X.Y.); (F.Z.); (B.Z.); (H.Q.); (Y.X.); (W.P.); (B.L.); (F.W.); (P.L.); (Y.S.); (A.Q.)
| | - Aibin Qu
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, and Beijing Key Laboratory of Environment and Aging, Beijing 100069, China; (X.Y.); (F.Z.); (B.Z.); (H.Q.); (Y.X.); (W.P.); (B.L.); (F.W.); (P.L.); (Y.S.); (A.Q.)
| | - Ling Zhang
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, and Beijing Key Laboratory of Environment and Aging, Beijing 100069, China; (X.Y.); (F.Z.); (B.Z.); (H.Q.); (Y.X.); (W.P.); (B.L.); (F.W.); (P.L.); (Y.S.); (A.Q.)
| |
Collapse
|
9
|
Lang J, Bernal A, Wist J, Egan S, Bong SH, Millet O, Ryan M, Lee AC, Hall D, Nitschke P, Masuda R, Imrie A, Holmes E, Nicholson J, Loo RL. Longitudinal study on immunologic, lipoproteomic, and inflammatory responses indicates the safety of sequential COVID-19 vaccination. J Mol Med (Berl) 2025; 103:421-433. [PMID: 40074874 PMCID: PMC12003606 DOI: 10.1007/s00109-025-02527-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 02/18/2025] [Accepted: 02/24/2025] [Indexed: 03/14/2025]
Abstract
COVID-19 vaccines are crucial in reducing SARS-CoV-2 transmission and severe health outcomes. Despite widespread administration, their long-term systemic effects on human metabolism remain inadequately understood. This longitudinal study aims to evaluate IgG responses, 34 cytokines, 112 lipoproteins, and 21 low-molecular-weight metabolites in 33 individuals receiving two to four COVID-19 vaccine doses. Changes in metabolic profiles for the first 16 days post each dose of vaccine, and up to 480 days post-initial dose, were compared to baseline (before vaccination). Additionally, metabolic profiles of vaccinated participants were compared to a reference cohort of unvaccinated individuals without prior exposure to SARS-CoV-2 infection (controls) and SARS-CoV-2 cases. Positive IgG responses were observed in 78.8% (N = 26) of participants after the first dose, reaching 100% with subsequent doses. The most common side effects were localized pain at the injection site and "flu-like" symptoms, reported by > 50% of participants. Systemic side effects, e.g., sore lymph nodes, fatigue, and brain fog, were reported but showed no significant correlations to IgG responses. Transient temporal changes were observed for cytokine IP10 (CXCL10) and glutamic acid around the third vaccine dose. Compared to the reference cohort, 497 vaccinated samples (95.0%) had profiles similar to the controls, while the remaining 26 samples with prior infection exposures were similar to mild cases of SARS-CooV-2 infection. In conclusion, COVID-19 vaccination did not induce lasting changes in inflammatory and metabolic responses, nor did it induce changes similar to mild cases of SARS-CoV-2 infection. This supports the metabolic safety of the vaccine and contributes to increased vaccine confidence. KEY MESSAGES: Minimal changes in inflammatory/metabolic markers up to 480 days post-vaccination. Transient increase in IP10 (CXCL10) and glutamic acid around the third dose. Post-vaccination IgG response did not alter metabolic profiles like SARS-CoV-2 cases. Our findings provide insights into the safety of repeated COVID-19 vaccinations.
Collapse
Affiliation(s)
- Jurissa Lang
- Australian National Phenome Centre and Centre for Computational and Systems Medicine, Health Futures Institute, Murdoch University, 5 Robin Warren Drive, Perth, WA, 6150, Australia
| | - Andres Bernal
- Australian National Phenome Centre and Centre for Computational and Systems Medicine, Health Futures Institute, Murdoch University, 5 Robin Warren Drive, Perth, WA, 6150, Australia
| | - Julien Wist
- Australian National Phenome Centre and Centre for Computational and Systems Medicine, Health Futures Institute, Murdoch University, 5 Robin Warren Drive, Perth, WA, 6150, Australia
- Chemistry Department, Universidad del Valle, Cali, 76001, Colombia
- Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, Sir Alexander Fleming Building, South Kensington, London, SW7 2AZ, UK
| | - Siobhon Egan
- Australian National Phenome Centre and Centre for Computational and Systems Medicine, Health Futures Institute, Murdoch University, 5 Robin Warren Drive, Perth, WA, 6150, Australia
| | - Sze How Bong
- Australian National Phenome Centre and Centre for Computational and Systems Medicine, Health Futures Institute, Murdoch University, 5 Robin Warren Drive, Perth, WA, 6150, Australia
| | - Oscar Millet
- Centro de Investigación Cooperativa en Biociencias -CIC bioGUNE, Precision Medicine and Metabolism Laboratory, Basque Research and Technology Alliance, Bizkaia Science and Technology Park, 48160, Derio, Spain
| | - Monique Ryan
- Australian National Phenome Centre and Centre for Computational and Systems Medicine, Health Futures Institute, Murdoch University, 5 Robin Warren Drive, Perth, WA, 6150, Australia
| | - Aude-Claire Lee
- Australian National Phenome Centre and Centre for Computational and Systems Medicine, Health Futures Institute, Murdoch University, 5 Robin Warren Drive, Perth, WA, 6150, Australia
| | - Drew Hall
- Australian National Phenome Centre and Centre for Computational and Systems Medicine, Health Futures Institute, Murdoch University, 5 Robin Warren Drive, Perth, WA, 6150, Australia
| | - Philipp Nitschke
- Australian National Phenome Centre and Centre for Computational and Systems Medicine, Health Futures Institute, Murdoch University, 5 Robin Warren Drive, Perth, WA, 6150, Australia
| | - Reika Masuda
- Australian National Phenome Centre and Centre for Computational and Systems Medicine, Health Futures Institute, Murdoch University, 5 Robin Warren Drive, Perth, WA, 6150, Australia
| | - Allison Imrie
- School of Biomedical Sciences, University of Western Australia, Nedlands, WA, 6009, Australia
| | - Elaine Holmes
- Australian National Phenome Centre and Centre for Computational and Systems Medicine, Health Futures Institute, Murdoch University, 5 Robin Warren Drive, Perth, WA, 6150, Australia
- Nutrition Research, Department of Metabolism, Nutrition and Reproduction, Faculty of Medicine, Imperial College London, Sir Alexander Fleming Building, London, SW7 2AZ, UK
| | - Jeremy Nicholson
- Australian National Phenome Centre and Centre for Computational and Systems Medicine, Health Futures Institute, Murdoch University, 5 Robin Warren Drive, Perth, WA, 6150, Australia.
- Nutrition Research, Department of Metabolism, Nutrition and Reproduction, Faculty of Medicine, Imperial College London, Sir Alexander Fleming Building, London, SW7 2AZ, UK.
- Institute of Global Health and Innovation, Imperial College London, Faculty Building South Kensington Campus, London, SW7 2AZ, UK.
| | - Ruey Leng Loo
- Australian National Phenome Centre and Centre for Computational and Systems Medicine, Health Futures Institute, Murdoch University, 5 Robin Warren Drive, Perth, WA, 6150, Australia.
| |
Collapse
|
10
|
Leite JSM, Vilas-Boas EA, Takahashi HK, Munhoz AC, Araújo LCC, Carvalho CR, Jr JD, Curi R, Carpinelli AR, Cruzat V. Liver lipid metabolism, oxidative stress, and inflammation in glutamine-supplemented ob/ob mice. J Nutr Biochem 2025; 138:109842. [PMID: 39824260 DOI: 10.1016/j.jnutbio.2025.109842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 12/21/2024] [Accepted: 01/13/2025] [Indexed: 01/20/2025]
Abstract
Glutamine availability may be reduced in chronic diseases, such as type 2 diabetes mellitus (T2DM)-induced by obesity. Herein, the antioxidant, anti-inflammatory and lipid metabolism effects of chronic oral glutamine supplementation in its free and dipeptide form were assessed in ob/ob mice. Adult male C57BL/6J ob/ob mice were supplemented with L-alanyl-L-glutamine (DIP) or free L-glutamine (GLN) in the drinking water for 40 days, whilst C57BL/6J Wild-type lean (WT) and control ob/ob mice (CTRL) received fresh water only. Plasma and tissue (skeletal muscle and liver) glutamine levels, and insulin resistance parameters (e.g., GTT, ITT, insulin) were determined. Oxidative stress (e.g., GSH system, Nrf2 translocation), inflammatory (e.g., NFkB translocation, TNF-α gene expression) and lipid metabolism parameters (e.g., plasma and liver triglyceride levels, SRBP-1, FAS, ACC, and ChRBP gene expression) were also analyzed. CTRL ob/ob mice showed lower glutamine levels in plasma and tissue, as well as increased insulin resistance and fat in the liver. Conversely, chronic DIP supplementation restored glutamine levels in plasma and tissues, improved glucose homeostasis and reduced plasma and liver lipid levels. Also, Nrf2 restoration, reduced NFkB translocation, and lower TNF-α gene expression was observed in the DIP group. Interestingly, chronic free GLN only increased muscle glutamine stores but reduced overall insulin resistance, and attenuated plasma and liver lipid metabolic biomarkers. The results presented herein indicate that restoration of body glutamine levels reduces oxidative stress and inflammation in obese and T2DM ob/ob mice. This effect attenuated hepatic lipid metabolic changes observed in obesity.
Collapse
Affiliation(s)
- Jaqueline Santos Moreira Leite
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, São Paulo, Brazil
| | - Eloisa Aparecida Vilas-Boas
- Department of Clinical and Toxicological Analysis, Faculty of Pharmaceutical Sciences, University of Sao Paulo, São Paulo, São Paulo, Brazil
| | - Hilton K Takahashi
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, São Paulo, Brazil
| | - Ana Cláudia Munhoz
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, São Paulo, Brazil
| | - Layanne C C Araújo
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, São Paulo, Brazil
| | - Carla Roberta Carvalho
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, São Paulo, Brazil
| | - Jose Donato Jr
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, São Paulo, Brazil
| | - Rui Curi
- Interdisciplinary Post-graduate Program in Health Sciences, ICAFE, Universidade Cruzeiro do Sul, São Paulo, São Paulo, Brazil; Instituto Butantan, São Paulo, São Paulo, Brazil
| | - Angelo Rafael Carpinelli
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, São Paulo, Brazil
| | - Vinicius Cruzat
- Faculty of Health, Southern Cross University, Gold Coast, Queensland, Australia.
| |
Collapse
|
11
|
Sarmento EB, Sassone LM, Pinto KP, Ferreira CMA, da Fidalgo TKS, Lopes RT, Alves ATNN, Freitas-Fernandes LB, Valente AP, Neves RH, da Silva EJNL. Evaluation of a potential bidirectional influence of metabolic syndrome and apical periodontitis: An animal-based study. Int Endod J 2025; 58:467-483. [PMID: 39797578 DOI: 10.1111/iej.14189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 11/24/2024] [Accepted: 12/23/2024] [Indexed: 01/13/2025]
Abstract
AIM This study aimed to explore the possible bidirectional interrelations between fructose-induced metabolic syndrome (MS) and apical periodontitis (AP). METHODOLOGY Twenty-eight male Wistar rats were distributed into four groups (n = 7, per group): Control (C), AP, Fructose Consumption (FRUT) and Fructose Consumption and AP (FRUT+AP). The rats in groups C and AP received filtered water, while those in groups FRUT and FRUT+AP received a 20% fructose solution mixed with water to induce MS. The groups AP and FRUT+AP had the pulp of their right mandibular first molar exposed to induce AP. Food consumption, murinometric measurements, blood glucose levels and glucose tolerance were monitored. Fifty-six days after the start of the experiment, the animals were euthanized, and serum samples were collected for metabolomic analysis. Mandibles, livers and right kidneys were also collected. The area and volume of the periapical lesions were calculated using micro-computed tomography. Histopathological evaluation was performed. Kruskal-Wallis followed by the Student-Newman-Keuls or Mann-Whitney tests and one-way anova followed by Tukey's or Independent t-test were used for non-parametric and parametric data, respectively (p < .05). Multivariate analysis and variable importance in projection score were applied to assess metabolite profile differences among groups (p < .05). RESULTS FRUT and FRUT+AP groups showed significantly increased fluid intake, body mass, abdominal circumference, blood glucose levels, liver weight and visceral fat weight (p < .05), indicating the development of MS. The analyses of the metabolite profile suggest increasing glucose, histidine, lactate, fatty acid and phenylalanine in the FRUT+AP group. There were no significant differences in volume and area of periapical lesions in micro-CT analyses (p = .1048 and p = .7494, respectively). Histopathological analysis of the hemimandibles demonstrated areas of inflammatory response, necrosis and microabscess in the periapical region. Hepatic histopathological observations indicated notable differences in cell appearance, with the FRUT and FRUT+AP groups showing signs of microsteatosis. Kidney analysis revealed Bowman's space dilation in the FRUT and AP groups, while the FRUT+AP group exhibited retracted Bowman's space, suggesting a possible alteration in renal filtration capacity. CONCLUSIONS MS had no impact on the progression of AP in rats. However, AP exacerbated the systemic state affected by MS, with changes in liver and kidney tissues and metabolite levels.
Collapse
Affiliation(s)
- Estéfano Borgo Sarmento
- Department of Integrated Clinical Procedures, School of Dentistry, Rio de Janeiro State University (UERJ), Rio de Janeiro, Brazil
| | - Luciana Moura Sassone
- Department of Integrated Clinical Procedures, School of Dentistry, Rio de Janeiro State University (UERJ), Rio de Janeiro, Brazil
| | - Karem Paula Pinto
- Department of Integrated Clinical Procedures, School of Dentistry, Rio de Janeiro State University (UERJ), Rio de Janeiro, Brazil
| | - Cláudio Malizia Alves Ferreira
- Department of Integrated Clinical Procedures, School of Dentistry, Rio de Janeiro State University (UERJ), Rio de Janeiro, Brazil
| | - Tatiana Kelly Silva da Fidalgo
- Department of Community and Preventive Dentistry, School of Dentistry, Rio de Janeiro State University (UERJ), Rio de Janeiro, Brazil
| | - Ricardo Tadeu Lopes
- Nuclear Engineering Program, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | | | - Liana Bastos Freitas-Fernandes
- National Center for Nuclear Magnetic Resonance, Medical Biochemistry, Rio de Janeiro Federal University, Rio de Janeiro, Brazil
| | - Ana Paula Valente
- National Center for Nuclear Magnetic Resonance, Medical Biochemistry, Rio de Janeiro Federal University, Rio de Janeiro, Brazil
| | - Renata Heisler Neves
- Romero Lascasas Porto Helminthology Laboratory, Department of Microbiology, Immunology and Parasitology, Medical Sciences College (FCM), Rio de Janeiro State University (UERJ), Rio de Janeiro, Brazil
| | - Emmanuel João Nogueira Leal da Silva
- Department of Integrated Clinical Procedures, School of Dentistry, Rio de Janeiro State University (UERJ), Rio de Janeiro, Brazil
- Department of Endodontics, Grande Rio University (UNIGRANRIO), Rio de Janeiro, Brazil
| |
Collapse
|
12
|
Chitteti R, Zuniga-Hertz JP, Masso-Silva JA, Shin J, Niesman I, Bojanowski CM, Kumar AJ, Hepokoski M, Crotty Alexander LE, Patel HH, Roth DM. E-cigarette-induced changes in cell stress and mitochondrial function. Free Radic Biol Med 2025; 228:329-338. [PMID: 39756490 DOI: 10.1016/j.freeradbiomed.2025.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 12/22/2024] [Accepted: 01/02/2025] [Indexed: 01/07/2025]
Abstract
Inhaling aerosols from electronic nicotine delivery systems, such as e-cigarettes (e-cigs), may pose health risks beyond those caused by nicotine intake. Exposure to e-cig aerosols can lead to the release of exosomes and metabolites into the bloodstream, potentially affecting mitochondrial physiology across the body, leading to chronic inflammatory diseases. In this study we assessed the effects of e-cig use by young healthy human subjects on the circulating exosome profile and markers of cell stress, and also defined the effects of e-cig user plasma on mitochondrial function in endothelial cells (EA. Hy 926) and epithelial cells (A549) via adoptive transfer. E-cig users had altered plasma exosome profiles, with significantly increased levels of cell free mitochondrial DNA (mtDNA), protein carbonyls, and 4-HNE relative to non-users. Plasma from e-cig users decreased maximal mitochondrial respiration and spare capacity of cells, while also increasing metabolic stress, as evidenced by changes in mitochondrial phenotype from basal to stressed in both endothelial and epithelial cells, which was corroborated by electron microscopy demonstrating structural changes in mitochondria. Mitochondrial membrane potential (MMP) and reactive oxygen species (ROS) levels significantly increased in e-cig plasma-subjected cells. Overall, we identified alterations in plasma exosome profiles and increased markers of mitochondrial stress in e-cig users and evidence that circulating factors within plasma from e-cig users drives metabolic stress in endothelial and epithelial cells. Our results imply that e-cig use adversely affects mitochondrial function, leading to stress and potentially chronic inflammation across the body.
Collapse
Affiliation(s)
- Ramamurthy Chitteti
- VA San Diego Healthcare System, San Diego, CA, USA; Department of Anesthesiology, School of Medicine, University of California San Diego, USA.
| | - Juan Pablo Zuniga-Hertz
- VA San Diego Healthcare System, San Diego, CA, USA; Department of Anesthesiology, School of Medicine, University of California San Diego, USA
| | - Jorge A Masso-Silva
- VA San Diego Healthcare System, San Diego, CA, USA; Division of Pulmonary, Critical Care and Sleep Medicine, University of California San Diego, USA
| | - John Shin
- Division of Pulmonary, Critical Care and Sleep Medicine, University of California San Diego, USA
| | - Ingrid Niesman
- San Diego State University, Electron Microscope Facility, 5500 Campanile Dr, San Diego, CA, 92182, USA
| | - Christine M Bojanowski
- Division of Pulmonary, Critical Care and Sleep Medicine, University of California San Diego, USA; Division of Pulmonary and Critical Care, Tulane University, New Orleans, LA, USA
| | - Avnee J Kumar
- VA San Diego Healthcare System, San Diego, CA, USA; Division of Pulmonary, Critical Care and Sleep Medicine, University of California San Diego, USA
| | - Mark Hepokoski
- VA San Diego Healthcare System, San Diego, CA, USA; Division of Pulmonary, Critical Care and Sleep Medicine, University of California San Diego, USA
| | - Laura E Crotty Alexander
- VA San Diego Healthcare System, San Diego, CA, USA; Division of Pulmonary, Critical Care and Sleep Medicine, University of California San Diego, USA
| | - Hemal H Patel
- VA San Diego Healthcare System, San Diego, CA, USA; Department of Anesthesiology, School of Medicine, University of California San Diego, USA
| | - David M Roth
- VA San Diego Healthcare System, San Diego, CA, USA; Department of Anesthesiology, School of Medicine, University of California San Diego, USA
| |
Collapse
|
13
|
Yang B, Li Z, Li P, Liang B, Liu Y, Feng E. Role of T cell metabolism in brain tumor development: a genetic and metabolic approach. BMC Neurol 2025; 25:12. [PMID: 39780065 PMCID: PMC11708232 DOI: 10.1186/s12883-024-04015-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 12/30/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Malignant brain tumors are among the most lethal cancers. Recent studies emphasized the crucial involvement of the immune system, especially T cells, in driving tumor progression and influencing patient outcomes. The emerging field of immunometabolism has shown that metabolic pathways play a pivotal role in regulating immune responses within the tumor microenvironment. This study aims to clarify the relationships between specific T cell phenotypes, circulating metabolites, and malignant brain tumors. METHODS We utilized a multiple mendelian randomization approach to investigate the associations between T cell phenotypes and malignant brain tumors, as well as the role of plasma metabolites in mediating these interactions. Instrumental variables were selected based on stringent criteria, and multiple mendelian randomization methods were utilized to identify causal pathways and metabolites potentially mediating these effects. RESULTS Our analysis identified significant associations between seven distinct T cell phenotypes, including various CD8 + and regulatory T cell subsets, and the presence of malignant brain tumors. We also identified 87 plasma metabolites correlated with these tumors. Notably, metabolites such as octadecanedioylcarnitine (C18-DC) and eicosanedioate (C20-DC) were implicated in modulating the risk of developing malignant brain tumors. Furthermore, metabolites such as 5-dodecenoate (12:1n7) and arachidonate (20:4n6) were found to influence tumor risk, particularly in relation to CD28 - CD8 + T cells. CONCLUSION The study identifies key T cell phenotypes and plasma metabolites involved in the pathogenesis of malignant brain tumors, offering potential biomarkers and therapeutic targets for future interventions.
Collapse
Affiliation(s)
- Bo Yang
- Department of Neurosurgery, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China
| | - Zhenyu Li
- Department of Neonatology , The First Hospital of Jilin University, Changchun, China
| | - Peiliang Li
- Department of Neurosurgery, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China
| | - Bo Liang
- Department of Neurosurgery, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China
| | - Yuhan Liu
- Department of Neurosurgery, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China
| | - Enshan Feng
- Department of Neurosurgery, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China.
| |
Collapse
|
14
|
Xie R, Herder C, Sha S, Peng L, Brenner H, Schöttker B. Novel type 2 diabetes prediction score based on traditional risk factors and circulating metabolites: model derivation and validation in two large cohort studies. EClinicalMedicine 2025; 79:102971. [PMID: 39720612 PMCID: PMC11667638 DOI: 10.1016/j.eclinm.2024.102971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 11/13/2024] [Accepted: 11/13/2024] [Indexed: 12/26/2024] Open
Abstract
Background We aimed to evaluate the incremental predictive value of metabolomic biomarkers for assessing the 10-year risk of type 2 diabetes when added to the clinical Cambridge Diabetes Risk Score (CDRS). Methods We utilized 86,232 UK Biobank (UKB) participants (recruited between 13 March 2006 and 1 October 2010) for model derivation and internal validation. Additionally, we included 4383 participants from the German ESTHER cohort (recruited between 1 July 2000 and 30 June 2002 for external validation). Participants were followed up for 10 years to assess the incidence of type 2 diabetes. A total of 249 NMR-derived metabolites were quantified using nuclear magnetic resonance (NMR) spectroscopy. Metabolites were selected with LASSO regression and model performance was evaluated with Harrell's C-index. Findings 11 metabolomic biomarkers, including glycolysis related metabolites, ketone bodies, amino acids, and lipids, were selected. In internal validation within the UKB, adding these metabolites significantly increased the C-index (95% confidence interval (95% CI)) of the clinical CDRS from 0.815 (0.800, 0.829) to 0.834 (0.820, 0.847) and the continuous net reclassification index (NRI) with 95% CI was 39.8% (34.6%, 45.0%). External validation in the ESTHER cohort showed a comparable statistically significant C-index increase from 0.770 (0.750, 0.791) to 0.798 (0.779, 0.817) and a continuous NRI of 33.8% (26.4%, 41.2%). A concise model with 4 instead of 11 metabolites yielded similar results. Interpretation Adding 11 metabolites to the clinical CDRS led to a novel type 2 diabetes prediction model, we called UK Biobank Diabetes Risk Score (UKB-DRS), substantially outperformed the clinical CDRS. The concise version with 4 metabolites performed comparably. As only very few clinical information and a blood sample are needed for the UKB-DRS, and as high-throughput NMR metabolomics are becoming increasingly available at low costs, these models have considerable potential for routine clinical application in diabetes risk assessment. Funding The ESTHER study was funded by grants from the Baden-Württemberg state Ministry of Science, Research and Arts (Stuttgart, Germany), the Federal Ministry of Education and Research (Berlin, Germany), the Federal Ministry of Family Affairs, Senior Citizens, Women and Youth (Berlin, Germany), and the Saarland State Ministry of Health, Social Affairs, Women and the Family (Saarbrücken, Germany). The UK Biobank project was established through collaboration between various entities including the Wellcome Trust, the Medical Research Council, Department of Health, Scottish Government, and the Northwest Regional Development Agency. Additional funding was provided by the Welsh Assembly Government, British Heart Foundation, Cancer Research UK, and Diabetes UK, with support from the National Health Service (NHS). The German Diabetes Center is funded by the German Federal Ministry of Health (Berlin, Germany) and the Ministry of Culture and Science of the state North Rhine-Westphalia (Düsseldorf, Germany) and receives additional funding from the German Federal Ministry of Education and Research (BMBF) through the German Center for Diabetes Research (DZD e.V.).
Collapse
Affiliation(s)
- Ruijie Xie
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center, Im Neuenheimer Feld 581, Heidelberg, 69120, Germany
- Faculty of Medicine, Heidelberg University, Heidelberg, 69115, Germany
| | - Christian Herder
- Institute for Clinical Diabetology, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Munich-Neuherberg, Germany
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Sha Sha
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center, Im Neuenheimer Feld 581, Heidelberg, 69120, Germany
- Faculty of Medicine, Heidelberg University, Heidelberg, 69115, Germany
| | - Lei Peng
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center, Im Neuenheimer Feld 581, Heidelberg, 69120, Germany
- Faculty of Medicine, Heidelberg University, Heidelberg, 69115, Germany
| | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center, Im Neuenheimer Feld 581, Heidelberg, 69120, Germany
- Network Aging Research, Heidelberg University, Heidelberg, Germany
| | - Ben Schöttker
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center, Im Neuenheimer Feld 581, Heidelberg, 69120, Germany
| |
Collapse
|
15
|
Peng M, Fu Y, Qin C, Shi L, Zhang M, Zhou S. A stratified study of human blood metabolites and coronary artery diseases-A Mendelian randomization study. Nutr Metab Cardiovasc Dis 2025; 35:103754. [PMID: 39448312 DOI: 10.1016/j.numecd.2024.09.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 09/07/2024] [Accepted: 09/23/2024] [Indexed: 10/26/2024]
Abstract
BACKGROUND AND AIMS Metabolic dysregulation is closely associated with coronary artery diseases (CAD). Exploring the relationship between metabolites and CAD is helpful in identifying changes in energy metabolism during disease progression. METHODS AND RESULTS We use Mendelian Randomization (MR) analysis to assess the relationships between 275 serum metabolites and CAD such as angina pectoris, post-myocardial infarction complications, coronary atherosclerosis, myocardial infarction (MI), and unstable angina pectoris (UA). The inverse variance-weighted method (IVW) served as the primary approach for causal analysis, with MR-Egger and weighted median (WM) as supplementary methods. Sensitivity analyses were conducted to assess heterogeneity and multiple effects. We also analyzed potentially related metabolic pathways.We identified causal relationships between 42 known metabolites and CAD. Among them, the genetic susceptibility to elevated levels of amino acid Isobutyrylcarnitine is associated with an increased risk of coronary artery atherosclerosis; but it provides protection against the development of MI. Genetic susceptibility to elevated levels of fatty acids Stearate, Caprylate is associated with higher risk of angina pectoris, while Threonate has a protective effect in the development of angina; Stearate is associated with an increased risk of UA, whereas higher levels of the lipids Choline, 1-arachidonoylglycerophosphoinositol∗, Hexadecanedioate, Tetradecanedioate play a protective role in UA.Metabolic pathway analysis identified 6 pathways that may be associated with CAD. CONCLUSION We identified causal relationships between 42 serum metabolites and CAD. Specifically, changes in metabolites such as Isobutyrylcarnitine, Caprylate, and Stearate were associated with risks of CAD. These findings provide new insights into the metabolic mechanisms of CAD.
Collapse
Affiliation(s)
- Mengling Peng
- The Center of Cardiovascular Diseases, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, Jilin, China
| | - Yu Fu
- The Center of Cardiovascular Diseases, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, Jilin, China
| | - Cong Qin
- The Center of Cardiovascular Diseases, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, Jilin, China
| | - Lei Shi
- The Center of Cardiovascular Diseases, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, Jilin, China
| | - Meiwei Zhang
- The Center of Cardiovascular Diseases, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, Jilin, China
| | - Shanshan Zhou
- The Center of Cardiovascular Diseases, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, Jilin, China.
| |
Collapse
|
16
|
Hunter B, Li M, Parker BL, Koay YC, Harney DJ, Pearson E, Cao J, Chen GT, Guneratne O, Smyth GK, Larance M, O'Sullivan JF, Lal S. Proteomic and metabolomic analyses of the human adult myocardium reveal ventricle-specific regulation in end-stage cardiomyopathies. Commun Biol 2024; 7:1666. [PMID: 39702518 DOI: 10.1038/s42003-024-07306-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 11/22/2024] [Indexed: 12/21/2024] Open
Abstract
The left and right ventricles of the human heart are functionally and developmentally distinct such that genetic or acquired insults can cause dysfunction in one or both ventricles resulting in heart failure. To better understand ventricle-specific molecular changes influencing heart failure development, we first performed unbiased quantitative mass spectrometry on pre-mortem non-diseased human myocardium to compare the metabolome and proteome between the normal left and right ventricles. Constituents of gluconeogenesis, glycolysis, lipogenesis, lipolysis, fatty acid catabolism, the citrate cycle and oxidative phosphorylation were down-regulated in the left ventricle, while glycogenesis, pyruvate and ketone metabolism were up-regulated. Inter-ventricular significance of these metabolic pathways was then found to be diminished within end-stage dilated cardiomyopathy and ischaemic cardiomyopathy, while heart failure-associated pathways were increased in the left ventricle relative to the right within ischaemic cardiomyopathy, such as fluid sheer-stress, increased glutamine-glutamate ratio, and down-regulation of contractile proteins, indicating a left ventricular pathological bias.
Collapse
Affiliation(s)
- Benjamin Hunter
- Precision Cardiovascular Laboratory, The University of Sydney, Sydney, NSW, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Mengbo Li
- Bioinformatics Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia
| | - Benjamin L Parker
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, VIC, Australia
| | - Yen Chin Koay
- Precision Cardiovascular Laboratory, The University of Sydney, Sydney, NSW, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- Heart Research Institute, Newtown, NSW, Australia
| | - Dylan J Harney
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, Sydney, NSW, Australia
| | - Evangeline Pearson
- Paediatric Oncology and Haematology, Oxford Children's Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, England
| | - Jacob Cao
- Central Clinical School, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Gavin T Chen
- Department of Cardiology, Royal Prince Alfred Hospital, Camperdown, NSW, Australia
| | - Oneka Guneratne
- Kolling Institute, Royal North Shore Hospital, and Charles Perkins Centre, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Gordon K Smyth
- Bioinformatics Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, VIC, Australia
- School of Mathematics and Statistics, The University of Melbourne, Parkville, VIC, Australia
| | - Mark Larance
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - John F O'Sullivan
- Precision Cardiovascular Laboratory, The University of Sydney, Sydney, NSW, Australia.
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia.
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.
- Heart Research Institute, Newtown, NSW, Australia.
- Central Clinical School, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.
- Department of Cardiology, Royal Prince Alfred Hospital, Camperdown, NSW, Australia.
- Faculty of Medicine, TU Dresden, Dresden, Germany.
| | - Sean Lal
- Precision Cardiovascular Laboratory, The University of Sydney, Sydney, NSW, Australia.
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia.
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.
- Central Clinical School, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.
- Department of Cardiology, Royal Prince Alfred Hospital, Camperdown, NSW, Australia.
- The Baird Institute for Applied Heart and Lung Surgical Research, Sydney, NSW, Australia.
| |
Collapse
|
17
|
Vo DK, Trinh KTL. Emerging Biomarkers in Metabolomics: Advancements in Precision Health and Disease Diagnosis. Int J Mol Sci 2024; 25:13190. [PMID: 39684900 DOI: 10.3390/ijms252313190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 12/01/2024] [Accepted: 12/06/2024] [Indexed: 12/18/2024] Open
Abstract
Metabolomics has come to the fore as an efficient tool in the search for biomarkers that are critical for precision health approaches and improved diagnostics. This review will outline recent advances in biomarker discovery based on metabolomics, focusing on metabolomics biomarkers reported in cancer, neurodegenerative disorders, cardiovascular diseases, and metabolic health. In cancer, metabolomics provides evidence for unique oncometabolites that are important for early disease detection and monitoring of treatment responses. Metabolite profiling for conditions such as neurodegenerative and mental health disorders can offer early diagnosis and mechanisms into the disease especially in Alzheimer's and Parkinson's diseases. In addition to these, lipid biomarkers and other metabolites relating to cardiovascular and metabolic disorders are promising for patient stratification and personalized treatment. The gut microbiome and environmental exposure also feature among the influential factors in biomarker discovery because they sculpt individual metabolic profiles, impacting overall health. Further, we discuss technological advances in metabolomics, current clinical applications, and the challenges faced by metabolomics biomarker validation toward precision medicine. Finally, this review discusses future opportunities regarding the integration of metabolomics into routine healthcare to enable preventive and personalized approaches.
Collapse
Affiliation(s)
- Dang-Khoa Vo
- College of Pharmacy, Gachon University, 191 Hambakmoe-ro, Yeonsu-gu, Incheon 21936, Republic of Korea
| | - Kieu The Loan Trinh
- BioNano Applications Research Center, Gachon University, 1342 Seongnam-daero, Sujeong-gu, Seongnam-si 13120, Gyeonggi-do, Republic of Korea
| |
Collapse
|
18
|
Wu Y, Avcilar-Kücükgöze I, Santovito D, Atzler D. Amino Acid Metabolism and Autophagy in Atherosclerotic Cardiovascular Disease. Biomolecules 2024; 14:1557. [PMID: 39766264 PMCID: PMC11673637 DOI: 10.3390/biom14121557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 11/29/2024] [Accepted: 12/04/2024] [Indexed: 01/11/2025] Open
Abstract
Cardiovascular disease is the most common cause of mortality globally, accounting for approximately one out of three deaths. The main underlying pathology is atherosclerosis, a dyslipidemia-driven, chronic inflammatory disease. The interplay between immune cells and non-immune cells is of great importance in the complex process of atherogenesis. During atheroprogression, intracellular metabolic pathways, such as amino acid metabolism, are master switches of immune cell function. Autophagy, an important stress survival mechanism involved in maintaining (immune) cell homeostasis, is crucial during the development of atherosclerosis and is strongly regulated by the availability of amino acids. In this review, we focus on the interplay between amino acids, especially L-leucine, L-arginine, and L-glutamine, and autophagy during atherosclerosis development and progression, highlighting potential therapeutic perspectives.
Collapse
Affiliation(s)
- Yuting Wu
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität München, 80336 Munich, Germany; (Y.W.); (I.A.-K.)
| | - Irem Avcilar-Kücükgöze
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität München, 80336 Munich, Germany; (Y.W.); (I.A.-K.)
- DZHK (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, 80336 Munich, Germany
| | - Donato Santovito
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität München, 80336 Munich, Germany; (Y.W.); (I.A.-K.)
- DZHK (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, 80336 Munich, Germany
- Institute for Genetic and Biomedical Research (IRGB), Unit of Milan, National Research Council, 20133 Milan, Italy
| | - Dorothee Atzler
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität München, 80336 Munich, Germany; (Y.W.); (I.A.-K.)
- DZHK (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, 80336 Munich, Germany
- Walter Straub Institute of Pharmacology and Toxicology, Ludwig-Maximilians-Universität München, 80336 Munich, Germany
| |
Collapse
|
19
|
Murcy F, Borowczyk C, Gourion-Arsiquaud S, Torrino S, Ouahrouche N, Barouillet T, Dussaud S, Couralet M, Vaillant N, Merlin J, Berquand A, Kaikkonen MU, McClelland RL, Tressel W, Stein J, Thorp EB, Bertero T, Barbry P, Bailly-Maitre B, Gautier EL, Karjalainen MK, Kettunen J, Duca L, Shea S, Yvan-Charvet L. GLS2 links glutamine metabolism and atherosclerosis by remodeling artery walls. NATURE CARDIOVASCULAR RESEARCH 2024; 3:1454-1467. [PMID: 39562782 DOI: 10.1038/s44161-024-00566-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 10/23/2024] [Indexed: 11/21/2024]
Abstract
Metabolic dysregulation, including perturbed glutamine-glutamate homeostasis, is common among patients with cardiovascular diseases, but the underlying mechanisms remain largely unknown. Using the human MESA cohort, here we show that plasma glutamine-glutamate ratio is an independent risk factor for carotid plaque progression. Mice deficient in glutaminase-2 (Gls2), the enzyme that mediates hepatic glutaminolysis, developed accelerated atherosclerosis and susceptibility to catastrophic cardiac events, while Gls2 overexpression partially protected from disease progression. High-throughput transcriptional profiling and high-resolution structural biology imaging of aortas showed that Gls2 deficiency perturbed extracellular matrix composition and increased vessel stiffness. This results from an imbalance of glutamine- and glutamate-dependent cross-linked proteins within atherosclerotic lesions and cellular remodeling of plaques. Thus, hepatic glutaminolysis functions as a potent regulator of glutamine homeostasis, which affects the aortic wall structure during atherosclerotic plaque progression.
Collapse
Grants
- ERC2016COG724838 EC | EU Framework Programme for Research and Innovation H2020 | H2020 Priority Excellent Science | H2020 European Research Council (H2020 Excellent Science - European Research Council)
- 25-CE14-Glutacare Agence Nationale de la Recherche (French National Research Agency)
- 75N92020D00001 NHLBI NIH HHS
- 75N92020D00004 NHLBI NIH HHS
- N01-HC-95164 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- 75N92020D00007 NHLBI NIH HHS
- N01-HC-95165 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- N01-HC-95166 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- N01-HC-95167 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- N01-HC-95169 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- 75N92020D00001 NHLBI NIH HHS
- 75N92020D00004 NHLBI NIH HHS
- 75N92020D00007 NHLBI NIH HHS
Collapse
Affiliation(s)
- Florent Murcy
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Fédération Hospitalo-Universitaire (FHU) Oncoage, IHU ResprERA Respiratory Health, Environment and Ageing (RespirERA), Nice, France
| | - Coraline Borowczyk
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Fédération Hospitalo-Universitaire (FHU) Oncoage, IHU ResprERA Respiratory Health, Environment and Ageing (RespirERA), Nice, France
| | | | - Stéphanie Torrino
- Université Côte d'Azur, CNRS, Inserm, IPMC, IHU-RespirERA, Valbonne, France
| | | | - Thibault Barouillet
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Fédération Hospitalo-Universitaire (FHU) Oncoage, IHU ResprERA Respiratory Health, Environment and Ageing (RespirERA), Nice, France
| | | | - Marie Couralet
- Université Côte d'Azur, CNRS, Inserm, IPMC, IHU-RespirERA, Valbonne, France
| | - Nathalie Vaillant
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Fédération Hospitalo-Universitaire (FHU) Oncoage, IHU ResprERA Respiratory Health, Environment and Ageing (RespirERA), Nice, France
| | - Johanna Merlin
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Fédération Hospitalo-Universitaire (FHU) Oncoage, IHU ResprERA Respiratory Health, Environment and Ageing (RespirERA), Nice, France
| | | | - Minna U Kaikkonen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | | | - William Tressel
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - James Stein
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Edward B Thorp
- Department of Pathology, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | - Thomas Bertero
- Université Côte d'Azur, CNRS, Inserm, IPMC, IHU-RespirERA, Valbonne, France
| | - Pascal Barbry
- Université Côte d'Azur, CNRS, Inserm, IPMC, IHU-RespirERA, Valbonne, France
| | - Béatrice Bailly-Maitre
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Fédération Hospitalo-Universitaire (FHU) Oncoage, IHU ResprERA Respiratory Health, Environment and Ageing (RespirERA), Nice, France
| | | | - Minna K Karjalainen
- Research Unit of Population Health, Faculty of Medicine, University of Oulu, Oulu, Finland
- Northern Finland Birth Cohorts, Arctic Biobank, Infrastructure for Population Studies, Faculty of Medicine, University of Oulu, Oulu, Finland
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Johannes Kettunen
- Research Unit of Population Health, Faculty of Medicine, University of Oulu, Oulu, Finland
- Biocenter Oulu, University of Oulu, Oulu, Finland
- Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
| | | | - Steven Shea
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Laurent Yvan-Charvet
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Fédération Hospitalo-Universitaire (FHU) Oncoage, IHU ResprERA Respiratory Health, Environment and Ageing (RespirERA), Nice, France.
| |
Collapse
|
20
|
Kaihara JNS, de Moraes FR, Nunes PR, Alves MG, Cavalli RC, Tasic L, Sandrim VC. Plasma metabolic profile reveals signatures of maternal health during gestational hypertension and preeclampsia without and with severe features. PLoS One 2024; 19:e0314053. [PMID: 39591465 PMCID: PMC11594399 DOI: 10.1371/journal.pone.0314053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 11/04/2024] [Indexed: 11/28/2024] Open
Abstract
Preeclampsia, a pregnancy-specific syndrome, poses substantial risks to maternal and neonatal health, particularly in cases with severe features. Our study focuses on evaluating the impact of low molecular weight metabolites on the intricate mechanisms and pathways involved in the pathophysiology of preeclampsia when severe features are present. We aim to pinpoint the distinct metabolomic profile in maternal plasma during pregnancies affected by hypertensive disorders and to correlate the metabolite levels with the clinical characteristics of the study cohort. A total of 173 plasma samples were collected, comprising 36 healthy pregnant women (HP), 52 patients with gestational hypertension (GH), 43 with preeclampsia without (PE-), and 42 with severe features (PE+). Nuclear magnetic resonance spectroscopy and metabolite identification were conducted to establish the metabolomic profiles. Univariate and chemometric analyses were conducted using MetaboAnalyst, and correlations were performed using GraphPad Prism. Our study unveils distinct metabolomic profiles differentiating HP women, patients featuring GH, and patients with PE-and PE+. Our analysis highlights an increase in acetate, N,N-dimethylglycine, glutamine, alanine, valine, and creatine levels in the PE+ group compared to the HP and GH groups. The PE+ group exhibited higher concentrations of N,N-dimethylglycine, glutamine, alanine, and valine compared to the PE-group. Moreover, elevated levels of specific metabolites, including N,N-dimethylglycine, alanine, and valine, were associated with increased blood pressure, worse obstetric outcomes, and poorer end-organ function, particularly renal and hepatic damage. Metabolomic analysis of PE+ individuals indicates heightened disturbances in nitrogen metabolism, methionine, and urea cycles. Additionally, the exacerbated metabolic disturbance may have disclosed renal impairment and hepatic dysfunction, evidenced by elevated levels of creatine and alanine. These findings not only contribute novel insights but also provide a more comprehensive understanding of the pathophysiological mechanisms at play in cases of PE+.
Collapse
Affiliation(s)
- Julyane N. S. Kaihara
- Department of Biophysics and Pharmacology, Institute of Biosciences of Botucatu, Sao Paulo State University (UNESP), Botucatu, SP, Brazil
| | - Fabio Rogerio de Moraes
- Multiuser Center for Biomolecular Innovation, Department of Physics, Institute of Biosciences, Languages and Exact Sciences, Sao Paulo State University (UNESP), Sao Jose do Rio Preto, SP, Brazil
| | - Priscila Rezeck Nunes
- Department of Biophysics and Pharmacology, Institute of Biosciences of Botucatu, Sao Paulo State University (UNESP), Botucatu, SP, Brazil
| | - Marco G. Alves
- Institute of Biomedicine and Department of Medical Science (iBiMED), University of Aveiro, Aveiro, Portugal
| | - Ricardo C. Cavalli
- Department of Gynecology and Obstetrics, Faculty of Medicine of Ribeirao Preto, University of Sao Paulo (USP), Ribeirao Preto, SP, Brazil
| | - Ljubica Tasic
- Department of Organic Chemistry, Institute of Chemistry, State University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Valeria Cristina Sandrim
- Department of Biophysics and Pharmacology, Institute of Biosciences of Botucatu, Sao Paulo State University (UNESP), Botucatu, SP, Brazil
| |
Collapse
|
21
|
Zhang Y, Qiu J, Sun S, Fang X. Altered amino acid levels in young hypopituitarism: impact of NAFLD and insulin resistance. Amino Acids 2024; 56:65. [PMID: 39580591 PMCID: PMC11585508 DOI: 10.1007/s00726-024-03429-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Accepted: 11/17/2024] [Indexed: 11/25/2024]
Abstract
Elevated concentrations of amino acids (AAs) are commonly observed in patients with nonalcoholic fatty liver disease (NAFLD). Individuals with hypopituitarism (HP) are at a heightened risk of developing NAFLD due to factors such as visceral obesity, increased insulin resistance (IR), and disturbances in lipid metabolism. However, the changes in AAs concentrations associated with HP remain poorly understood. Therefore, our study aimed to investigate whether individuals with HP, who were not receiving growth hormone replacement therapy (GHRT), exhibited altered AAs compared to controls (CTs), and whether these AAs were associated with IR, the presence of NAFLD, and the Metabolic Syndrome (MetS) score. The AAs profiles of 133 young males with HP (age: 24.5 ± 5.9; 57 with NAFLD and 76 without NAFLD) and 90 age and BMI-matched CTs were analyzed using untargeted metabolomics. The results revealed that most AAs were found to be elevated in subjects with HPs compared to CTs. Glutamate, glutamine, norleucine, and branched-chain amino acids (BCAAs) (leucine and valine) were correlated with the homeostasis model assessment of insulin resistance (HOMA-IR), with glutamate and norleucine showing independent linkage. Glutamate and proline levels were specifically associated with MetS score, while alanine and proline linked to NAFLD. Given that elevated glutamate and BCAAs levels have higher prevalence of NAFLD, we hypothesized that the changes in AAs observed in HPs may be attributed to the impact of NAFLD and IR.
Collapse
Affiliation(s)
- Yuwen Zhang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jiting Qiu
- Department of Neurosurgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Shouyue Sun
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Xuqian Fang
- Department of Pathology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
22
|
Prechtl L, Carrard J, Gallart-Ayala H, Borreggine R, Teav T, Königstein K, Wagner J, Knaier R, Infanger D, Streese L, Hinrichs T, Hanssen H, Ivanisevic J, Schmidt-Trucksäss A. Circulating amino acid signature features urea cycle alterations associated with coronary artery disease. Sci Rep 2024; 14:25848. [PMID: 39468229 PMCID: PMC11519371 DOI: 10.1038/s41598-024-76835-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 10/17/2024] [Indexed: 10/30/2024] Open
Abstract
Coronary artery disease (CAD) remains a leading cause of death worldwide and imposes a substantial socioeconomic burden on healthcare. Improving risk stratification in clinical practice could help to combat this burden. As amino acids are biologically active metabolites whose involvement in CAD remains largely unknown, this study investigated associations between circulating amino acid levels and CAD phenotypes. A high-coverage quantitative liquid chromatography-mass spectrometry approach was applied to acquire the serum amino acids profile of age- and sex-coarsened-matched patients with CAD (n = 46, 66.9 years, 74.7% male) and healthy individuals (n = 120, 67.4 years, 74.7% male) from the COmPLETE study. Multiple linear regressions were performed to investigate associations between amino acid levels and (a) the health status (CAD vs. healthy), (b) the number of affected coronary arteries, or (c) the left ventricular ejection fraction. Regressions were adjusted for age, sex, daily physical activity, sampling, and fasting time. Urea cycle amino acids (ornithine, citrulline, homocitrulline, aspartate, and arginine) were significantly and negatively associated with CAD, the number of affected coronary arteries, and the left ventricular ejection fraction. Lysine, histidine, and the glutamine/glutamate ratio were also significantly and negatively associated with the CAD phenotypes. Overall, patients with CAD displayed lower levels of urea cycle amino acids, highlighting a potential role for urea cycle amino acid profiling in cardiovascular risk stratification.Trial registrationThe study was registered on https://www.clinicaltrials.gov (NCT03986892) on June 5, 2019.
Collapse
Affiliation(s)
- Luisa Prechtl
- School of Cardiovascular and Metabolic Health, University of Glasgow, 126 University Place, Glasgow, G12 8TA, Scotland
| | - Justin Carrard
- Division of Sports and Exercise Medicine, Department of Sport, Exercise and Health, University of Basel, Grosse Allee 6, 4052, Basel, Switzerland.
| | - Hector Gallart-Ayala
- Metabolomics Platform, Faculty of Biology and Medicine, University of Lausanne, Quartier UNIL-CHUV-Rue du Bugnon 19, 1005, Lausanne, Switzerland
| | - Rébecca Borreggine
- Metabolomics Platform, Faculty of Biology and Medicine, University of Lausanne, Quartier UNIL-CHUV-Rue du Bugnon 19, 1005, Lausanne, Switzerland
| | - Tony Teav
- Metabolomics Platform, Faculty of Biology and Medicine, University of Lausanne, Quartier UNIL-CHUV-Rue du Bugnon 19, 1005, Lausanne, Switzerland
| | - Karsten Königstein
- Division of Sports and Exercise Medicine, Department of Sport, Exercise and Health, University of Basel, Grosse Allee 6, 4052, Basel, Switzerland
| | - Jonathan Wagner
- Division of Sports and Exercise Medicine, Department of Sport, Exercise and Health, University of Basel, Grosse Allee 6, 4052, Basel, Switzerland
| | - Raphael Knaier
- Division of Sports and Exercise Medicine, Department of Sport, Exercise and Health, University of Basel, Grosse Allee 6, 4052, Basel, Switzerland
| | - Denis Infanger
- Division of Sports and Exercise Medicine, Department of Sport, Exercise and Health, University of Basel, Grosse Allee 6, 4052, Basel, Switzerland
| | - Lukas Streese
- Division of Sports and Exercise Medicine, Department of Sport, Exercise and Health, University of Basel, Grosse Allee 6, 4052, Basel, Switzerland
| | - Timo Hinrichs
- Division of Sports and Exercise Medicine, Department of Sport, Exercise and Health, University of Basel, Grosse Allee 6, 4052, Basel, Switzerland
| | - Henner Hanssen
- Division of Sports and Exercise Medicine, Department of Sport, Exercise and Health, University of Basel, Grosse Allee 6, 4052, Basel, Switzerland
| | - Julijana Ivanisevic
- Metabolomics Platform, Faculty of Biology and Medicine, University of Lausanne, Quartier UNIL-CHUV-Rue du Bugnon 19, 1005, Lausanne, Switzerland.
| | - Arno Schmidt-Trucksäss
- Division of Sports and Exercise Medicine, Department of Sport, Exercise and Health, University of Basel, Grosse Allee 6, 4052, Basel, Switzerland
| |
Collapse
|
23
|
Han F, Xu C, Hangfu X, Liu Y, Zhang Y, Sun B, Chen L. Circulating glutamine/glutamate ratio is closely associated with type 2 diabetes and its associated complications. Front Endocrinol (Lausanne) 2024; 15:1422674. [PMID: 39092282 PMCID: PMC11291334 DOI: 10.3389/fendo.2024.1422674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 07/09/2024] [Indexed: 08/04/2024] Open
Abstract
Objective This study aims to conduct a comprehensive investigation of the serum amino acid profiles of individuals with type 2 diabetes (T2D) and its related complications. Methods Patients with T2D were enrolled in this study. Sixteen kinds of common amino acids in the fasting circulating were assessed through liquid chromatography-mass spectrometry (LC-MS). Subsequently, correlation, regression analyses, and receiver operating characteristic (ROC) curves were conducted to assess the associations between amino acids and clinical indicators. Results Thirteen different kinds of amino acids were identified in diabetic patients, as compared with normal controls. The Glutamine/Glutamate (Gln/Glu) ratio was negatively correlated with BMI, HbA1c, serum uric acid, and the triglyceride-glucose (TyG) index, while it was positively correlated with HDL-C. Logistic regression analyses indicated that Gln/Glu was a consistent protective factor for both T2D (OR = 0.65, 95% CI 0.50-0.86) and obesity (OR = 0.79, 95% CI 0.66-0.96). The ROC curves demonstrated that Gln/Glu, proline, valine, and leucine provided effective predictions for diabetes risk, with Gln/Glu exhibiting the highest AUC [0.767 (0.678-0.856)]. In patients with T2D, Gln was the only amino acid that displayed a negative correlation with HbA1c (r = -0.228, p = 0.017). Furthermore, HOMA-β exhibited a negative correlation with Glu (r = -0.301, p = 0.003) but a positive correlation with Gln/Glu (r = 0.245, p = 0.017). Notably, logistic regression analyses revealed an inverse correlation of Gln/Glu with the risk of diabetic kidney disease (OR = 0.74, 95% CI 0.55-0.98) and a positive association with the risk of diabetic retinopathy (OR = 1.53, 95% CI 1.08-2.15). Conclusion The Gln/Glu ratio exhibited a significant association with diabetes, common metabolic parameters, and diabetic complications. These findings shed light on the pivotal role of Gln metabolism in T2D and its associated complications.
Collapse
Affiliation(s)
| | | | | | | | | | - Bei Sun
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - Liming Chen
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| |
Collapse
|
24
|
Majdizadeh G, Beytollahi M, Djazayery A, Movahedi A. Role of Branched and Aromatic Amino Acids, Diet Inflammatory Index, and Anthropometric Indices on Mental Health. Int J Prev Med 2024; 15:23. [PMID: 39170923 PMCID: PMC11338367 DOI: 10.4103/ijpvm.ijpvm_59_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 09/13/2023] [Indexed: 08/23/2024] Open
Abstract
Background Mental health disorders are one of the most important and increasing health problems in the youth of today's societies. Some dietary intake and body mass status are factors that affect mental health. This study aimed to investigate the relationship between the intake of branched-chain amino acids (BCAAs) and aromatic amino acids (AAAs) and anthropometric and dietary inflammatory indices with mental health, including depression, anxiety, and stress. Methods In this case-control study, the data of 138 teenage girls aged 13-18 years were collected. Three-day 24-hour food recall and standard anthropometric methods were used to calculate the dietary inflammation intake score of normal and energy-adjusted diets. Mental health disorders were diagnosed by the DASS-21 questionnaire. Statistical analysis used Student's t-test, correlation, and multiple regression were used to analyze the data based on the study's statistical requirements. Results Based on the findings, 59 (42%) of the girls had mental disorders, and 79 (58%) were healthy. The average weight of stressed people was significantly higher than that of healthy people, and the BMI of anxious people was significantly higher than that of nonanxious people (P < 0.05). A significant positive correlation was found between stress and weight and energy intake. Additionally, there was a significant negative correlation between BCAAs and mental health. The average intake of BCAAs was significantly lower in patients (P = 0.01). The trend analysis showed significantly lower BCAA levels among the 4th quartile of mental disorders. No significant relationship was observed between DII, AAA, and anthropometric indices. After adjustment of the results, no relationship was observed between mental health and the studied factors. Conclusions BCAA might be related to mental health. Further studies in different age and sex groups are highly recommended.
Collapse
Affiliation(s)
- Golnaz Majdizadeh
- Department of Nutrition, Science and Research Branch Islamic Azad University, Tehran, Iran
| | - Mina Beytollahi
- Department of Nutrition, Science and Research Branch Islamic Azad University, Tehran, Iran
| | - Abolghasem Djazayery
- Department of Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Ariyo Movahedi
- Department of Nutrition, Science and Research Branch Islamic Azad University, Tehran, Iran
| |
Collapse
|
25
|
Liu N, Yin Z, Wang M, Kui H, Yuan Z, Tian Y, Liu C, Huang J. Pharmacodynamic and targeted amino acid metabolomics researches on the improvement of diabetic retinopathy with Fufang Xueshuantong component compatibility. J Chromatogr B Analyt Technol Biomed Life Sci 2024; 1242:124194. [PMID: 38924945 DOI: 10.1016/j.jchromb.2024.124194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 03/28/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024]
Abstract
The Fufang Xueshuantong capsule (FXT) has significant preventive and therapeutic effects on diabetic retinopathy(DR), but the compatibility of its active components remains to be thoroughly explored. In this study, a zebrafish diabetic retinopathy model was established using high-mixed sugars, and the optimal ratios of notoginseng total saponins, total salvianolic acid, astragaloside, and harpagide were selected through orthogonal experiments. Furthermore, we used UPLC-QqQ/MS to detect the changes in amino acid content of DR zebrafish tissues after administration of FXT and its compatible formula to analyze the effects of FXT and its compatible formula on amino acid metabolites. The results showed that the final compatibility ratios of the components were 8: 5: 1: 6.6 by comprehensive evaluation of the indicators. FXT and its compatibility formula had beneficial effects on retinal vasodilatation, lipid accumulation in the liver, total glucose, and VEGF levels in DR zebrafish, and all of them could call back some amino acid levels in DR zebrafish. In this research, we determined the compatible formulation of the active ingredients in the FXT and investigated their efficacy in DR zebrafish for further clinical applications.
Collapse
Affiliation(s)
- Ning Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China; Beijing Key Lab for Quality Evaluation of Chinese Materia Medica, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Ziqiang Yin
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Mingshuang Wang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
| | - Hongqian Kui
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Zhenshuang Yuan
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yue Tian
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Chuanxin Liu
- Luoyang Key Laboratory of Clinical Multiomics and Translational Medicine, Henan Key Laboratory of Rare Diseases, Endocrinology and Metabolism Center, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China, 471003.
| | - Jianmei Huang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China.
| |
Collapse
|
26
|
Mires S, Sommella E, Merciai F, Salviati E, Caponigro V, Basilicata MG, Marini F, Campiglia P, Baquedano M, Dong T, Skerritt C, Eastwood KA, Caputo M. Plasma metabolomic and lipidomic profiles accurately classify mothers of children with congenital heart disease: an observational study. Metabolomics 2024; 20:70. [PMID: 38955892 PMCID: PMC11219374 DOI: 10.1007/s11306-024-02129-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 05/08/2024] [Indexed: 07/04/2024]
Abstract
INTRODUCTION Congenital heart disease (CHD) is the most common congenital anomaly, representing a significant global disease burden. Limitations exist in our understanding of aetiology, diagnostic methodology and screening, with metabolomics offering promise in addressing these. OBJECTIVE To evaluate maternal metabolomics and lipidomics in prediction and risk factor identification for childhood CHD. METHODS We performed an observational study in mothers of children with CHD following pregnancy, using untargeted plasma metabolomics and lipidomics by ultrahigh performance liquid chromatography-high resolution mass spectrometry (UHPLC-HRMS). 190 cases (157 mothers of children with structural CHD (sCHD); 33 mothers of children with genetic CHD (gCHD)) from the children OMACp cohort and 162 controls from the ALSPAC cohort were analysed. CHD diagnoses were stratified by severity and clinical classifications. Univariate, exploratory and supervised chemometric methods were used to identify metabolites and lipids distinguishing cases and controls, alongside predictive modelling. RESULTS 499 metabolites and lipids were annotated and used to build PLS-DA and SO-CovSel-LDA predictive models to accurately distinguish sCHD and control groups. The best performing model had an sCHD test set mean accuracy of 94.74% (sCHD test group sensitivity 93.33%; specificity 96.00%) utilising only 11 analytes. Similar test performances were seen for gCHD. Across best performing models, 37 analytes contributed to performance including amino acids, lipids, and nucleotides. CONCLUSIONS Here, maternal metabolomic and lipidomic analysis has facilitated the development of sensitive risk prediction models classifying mothers of children with CHD. Metabolites and lipids identified offer promise for maternal risk factor profiling, and understanding of CHD pathogenesis in the future.
Collapse
Affiliation(s)
- Stuart Mires
- Translational Health Sciences, University of Bristol, Bristol, UK.
- University Hospitals Bristol and Weston NHS Foundation Trust, Bristol, UK.
| | | | | | | | - Vicky Caponigro
- Department of Pharmacy, University of Salerno, Salerno, Italy
| | - Manuela Giovanna Basilicata
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | | | | | - Mai Baquedano
- Translational Health Sciences, University of Bristol, Bristol, UK
| | - Tim Dong
- Translational Health Sciences, University of Bristol, Bristol, UK
| | - Clare Skerritt
- University Hospitals Bristol and Weston NHS Foundation Trust, Bristol, UK
| | - Kelly-Ann Eastwood
- Translational Health Sciences, University of Bristol, Bristol, UK
- University Hospitals Bristol and Weston NHS Foundation Trust, Bristol, UK
| | - Massimo Caputo
- Translational Health Sciences, University of Bristol, Bristol, UK
- University Hospitals Bristol and Weston NHS Foundation Trust, Bristol, UK
| |
Collapse
|
27
|
Lecoutre S, Maqdasy S, Rizo-Roca D, Renzi G, Vlassakev I, Alaeddine LM, Higos R, Jalkanen J, Zhong J, Zareifi DS, Frendo-Cumbo S, Massier L, Hodek O, Juvany M, Moritz T, de Castro Barbosa T, Omar-Hmeadi M, López-Yus M, Merabtene F, Abatan JB, Marcelin G, El Hachem EJ, Rouault C, Bergo MO, Petrus P, Zierath JR, Clément K, Krook A, Mejhert N, Rydén M. Reduced adipocyte glutaminase activity promotes energy expenditure and metabolic health. Nat Metab 2024; 6:1329-1346. [PMID: 39009762 PMCID: PMC11272588 DOI: 10.1038/s42255-024-01083-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 06/14/2024] [Indexed: 07/17/2024]
Abstract
Glutamine and glutamate are interconverted by several enzymes and alterations in this metabolic cycle are linked to cardiometabolic traits. Herein, we show that obesity-associated insulin resistance is characterized by decreased plasma and white adipose tissue glutamine-to-glutamate ratios. We couple these stoichiometric changes to perturbed fat cell glutaminase and glutamine synthase messenger RNA and protein abundance, which together promote glutaminolysis. In human white adipocytes, reductions in glutaminase activity promote aerobic glycolysis and mitochondrial oxidative capacity via increases in hypoxia-inducible factor 1α abundance, lactate levels and p38 mitogen-activated protein kinase signalling. Systemic glutaminase inhibition in male and female mice, or genetically in adipocytes of male mice, triggers the activation of thermogenic gene programs in inguinal adipocytes. Consequently, the knockout mice display higher energy expenditure and improved glucose tolerance compared to control littermates, even under high-fat diet conditions. Altogether, our findings highlight white adipocyte glutamine turnover as an important determinant of energy expenditure and metabolic health.
Collapse
Affiliation(s)
- Simon Lecoutre
- Department of Medicine (Huddinge), Karolinska Institutet, ME Endokrinologi, Karolinska University Hospital Huddinge, Huddinge, Sweden
- Nutrition and Obesities: Systemic Approaches Research Group, NutriOmics, Sorbonne Université, INSERM, Paris, France
| | - Salwan Maqdasy
- Department of Medicine (Huddinge), Karolinska Institutet, ME Endokrinologi, Karolinska University Hospital Huddinge, Huddinge, Sweden
| | - David Rizo-Roca
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Gianluca Renzi
- Department of Medicine (Huddinge), Karolinska Institutet, ME Endokrinologi, Karolinska University Hospital Huddinge, Huddinge, Sweden
| | - Ivan Vlassakev
- Department of Medicine (Huddinge), Karolinska Institutet, ME Endokrinologi, Karolinska University Hospital Huddinge, Huddinge, Sweden
| | - Lynn M Alaeddine
- Department of Medicine (Huddinge), Karolinska Institutet, ME Endokrinologi, Karolinska University Hospital Huddinge, Huddinge, Sweden
| | - Romane Higos
- Department of Medicine (Huddinge), Karolinska Institutet, ME Endokrinologi, Karolinska University Hospital Huddinge, Huddinge, Sweden
| | - Jutta Jalkanen
- Department of Medicine (Huddinge), Karolinska Institutet, ME Endokrinologi, Karolinska University Hospital Huddinge, Huddinge, Sweden
| | - Jiawei Zhong
- Department of Medicine (Huddinge), Karolinska Institutet, ME Endokrinologi, Karolinska University Hospital Huddinge, Huddinge, Sweden
| | - Danae S Zareifi
- Department of Medicine (Huddinge), Karolinska Institutet, ME Endokrinologi, Karolinska University Hospital Huddinge, Huddinge, Sweden
| | - Scott Frendo-Cumbo
- Department of Medicine (Huddinge), Karolinska Institutet, ME Endokrinologi, Karolinska University Hospital Huddinge, Huddinge, Sweden
| | - Lucas Massier
- Department of Medicine (Huddinge), Karolinska Institutet, ME Endokrinologi, Karolinska University Hospital Huddinge, Huddinge, Sweden
| | - Ondrej Hodek
- Swedish Metabolomics Centre, Department of Forest Genetics and Plant Physiology, Swedish University of Agricultural Sciences, Umeå, Sweden
| | - Marta Juvany
- Swedish Metabolomics Centre, Department of Forest Genetics and Plant Physiology, Swedish University of Agricultural Sciences, Umeå, Sweden
| | - Thomas Moritz
- Swedish Metabolomics Centre, Department of Forest Genetics and Plant Physiology, Swedish University of Agricultural Sciences, Umeå, Sweden
- The Novo Nordisk Foundation Centre for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Thais de Castro Barbosa
- Department of Medicine (Huddinge), Karolinska Institutet, ME Endokrinologi, Karolinska University Hospital Huddinge, Huddinge, Sweden
| | - Muhmmad Omar-Hmeadi
- Department of Medicine (Huddinge), Karolinska Institutet, ME Endokrinologi, Karolinska University Hospital Huddinge, Huddinge, Sweden
| | - Marta López-Yus
- Adipocyte and Fat Biology Laboratory (AdipoFat), Translational Research Unit, University Hospital Miguel Servet, Zaragoza, Spain
- Instituto Aragonés de Ciencias de La Salud (IACS), Zaragoza, Spain
- Instituto de Investigación Sanitaria (IIS)-Aragón, Zaragoza, Spain
| | - Fatiha Merabtene
- Nutrition and Obesities: Systemic Approaches Research Group, NutriOmics, Sorbonne Université, INSERM, Paris, France
| | - Jimon Boniface Abatan
- Nutrition and Obesities: Systemic Approaches Research Group, NutriOmics, Sorbonne Université, INSERM, Paris, France
| | - Geneviève Marcelin
- Nutrition and Obesities: Systemic Approaches Research Group, NutriOmics, Sorbonne Université, INSERM, Paris, France
| | - Elie-Julien El Hachem
- Nutrition and Obesities: Systemic Approaches Research Group, NutriOmics, Sorbonne Université, INSERM, Paris, France
| | - Christine Rouault
- Nutrition and Obesities: Systemic Approaches Research Group, NutriOmics, Sorbonne Université, INSERM, Paris, France
| | - Martin O Bergo
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - Paul Petrus
- Department of Medicine (Huddinge), Karolinska Institutet, ME Endokrinologi, Karolinska University Hospital Huddinge, Huddinge, Sweden
| | - Juleen R Zierath
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Karine Clément
- Nutrition and Obesities: Systemic Approaches Research Group, NutriOmics, Sorbonne Université, INSERM, Paris, France
- Nutrition Department, Assistance Publique Hôpitaux de Paris, CRNH Ile-de-France, Pitié-Salpêtrière Hospital, Paris, France
| | - Anna Krook
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Niklas Mejhert
- Department of Medicine (Huddinge), Karolinska Institutet, ME Endokrinologi, Karolinska University Hospital Huddinge, Huddinge, Sweden.
| | - Mikael Rydén
- Department of Medicine (Huddinge), Karolinska Institutet, ME Endokrinologi, Karolinska University Hospital Huddinge, Huddinge, Sweden.
| |
Collapse
|
28
|
Niu R, Wang H, Peng R, Wang W, Lin Y, Xiao Y, Zhou L, Xu X, Mu X, Zhang X, He M, Li W, Wu T, Qiu G. Associations of Plasma Metabolites With Risks of Incident Stroke and Its Subtypes in Chinese Adults. J Am Heart Assoc 2024; 13:e033201. [PMID: 38844434 PMCID: PMC11255744 DOI: 10.1161/jaha.123.033201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 05/13/2024] [Indexed: 06/19/2024]
Abstract
BACKGROUND Metabolomics studies have identified various metabolic markers associated with stroke risk, yet much uncertainty persists regarding heterogeneity in these associations between different stroke subtypes. We aimed to examine metabolic profiles associated with incident stroke and its subtypes in Chinese adults. METHODS AND RESULTS We performed a nested case-control study within the Dongfeng-Tongji cohort, including 1029 and 266 incident cases of ischemic stroke (IS) and hemorrhagic stroke (HS), respectively, with a mean follow-up period of 6.1±2.3 years. Fifty-five metabolites in fasting plasma were measured by ultra-high-performance liquid chromatography-mass spectrometry. We examined the associations of metabolites with the risks of total stroke, IS, and HS, with a focus on the comparison of associations of plasma metabolite with IS and HS, using conditional logistic regression. We found that increased levels of asymmetrical/symmetrical dimethylarginine and glutamate were significantly associated with elevated risk of total stroke (odds ratios and 95%, 1.20 [1.08-1.34] and 1.22 [1.09-1.36], respectively; both Benjamini-Hochberg-adjusted P <0.05). When examining stroke subtypes, asymmetrical/symmetrical dimethylarginine was nominally associated with both IS and HS (odds ratios [95% CIs]: 1.16 [1.03-1.31] and 1.39 [1.07-1.81], respectively), while glutamate was associated with only IS (odds ratios [95% CI]: 1.26 [1.11-1.43]). The associations of glutamate with IS risk were significantly stronger among participants with hypertension and diabetes than among those without these diseases (both P for interaction <0.05). CONCLUSIONS This study validated the positive associations of asymmetrical/symmetrical dimethylarginine and glutamate with stroke risk, mainly that of IS, in a Chinese population, and revealed a novel unanimous association of with both IS and HS. Our findings provided potential intervention targets for stroke prevention.
Collapse
Affiliation(s)
- Rundong Niu
- Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Hao Wang
- Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Rong Peng
- Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Wei Wang
- Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Yuhui Lin
- Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Yang Xiao
- Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Lue Zhou
- Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Xuedan Xu
- Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Xuanwen Mu
- Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Xiaomin Zhang
- Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Meian He
- Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Wending Li
- Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Department of Environmental Health SciencesMailman School of Public HealthColumbia UniversityNew YorkNYUSA
| | - Tangchun Wu
- Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Gaokun Qiu
- Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| |
Collapse
|
29
|
Han SK, Seo MJ, Lee T, Kim MY. Effectiveness of the ALT/AST ratio for predicting insulin resistance in a Korean population: A large-scale, cross-sectional cohort study. PLoS One 2024; 19:e0303333. [PMID: 38758828 PMCID: PMC11101110 DOI: 10.1371/journal.pone.0303333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 04/23/2024] [Indexed: 05/19/2024] Open
Abstract
Insulin resistance is a common pathophysiology in patients with type 2 diabetes mellitus, cardiovascular disease, and non-alcoholic fatty liver disease. Thus, screening for the risk of insulin resistance is important to prevent disease progression. We evaluated the alanine aminotransferase/aspartate aminotransferase (ALT/AST) ratio to predict insulin resistance in the general population, regardless of comorbidities. Datasets from the 2015, 2019, and 2020 Korea National Health and Nutrition Examination Surveys were used, and the following four indices were implemented to indicate insulin resistance: fasting serum glucose, insulin, homeostatic model assessment for insulin resistance (HOMA-IR), and β-cell function. We analyzed the degree of association between the liver enzyme profile and insulin resistance indices using Pearson's correlation coefficient and determined the associations using linear or logistic regression analysis. Accordingly, ALT levels in both sexes were positively and consistently correlated with the four aforementioned insulin resistance indices in stratification analyses based on diabetes, dyslipidemia, alcohol consumption, and obesity status. In multivariate linear regression, when comparing with ALT levels, the ALT/AST ratio exhibited superior predictive performance for fasting serum glucose and HOMA-β in Korean men and improved outcomes for all insulin resistance indices in Korean women. In this analysis that included a large community-based population, the ALT/AST ratio was a more useful predictive marker than the HOMA-IR. Regarding the predicted presence or absence of insulin resistance, the ALT/AST ratio could better predict HOMA-IR than the ALT level alone in Koreans. A simple, precise marker that represents the ALT/AST ratio could be a practical method to screen for insulin resistance in the general population, regardless of diabetes mellitus, alcohol intake, and sex.
Collapse
Affiliation(s)
- Seul Ki Han
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Yonsei University Wonju College of Medicine, Wonju, Korea
- Regeneration Medicine Research Center, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Myung Jae Seo
- Regeneration Medicine Research Center, Yonsei University Wonju College of Medicine, Wonju, Korea
- Department of Family Medicine, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Taesic Lee
- Regeneration Medicine Research Center, Yonsei University Wonju College of Medicine, Wonju, Korea
- Department of Family Medicine, Yonsei University Wonju College of Medicine, Wonju, Korea
- Division of Data Mining and Computational Biology, Institute of Global Health Care and Development, Wonju, Korea
| | - Moon Young Kim
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Yonsei University Wonju College of Medicine, Wonju, Korea
- Regeneration Medicine Research Center, Yonsei University Wonju College of Medicine, Wonju, Korea
| |
Collapse
|
30
|
Verkerke ARP, Wang D, Yoshida N, Taxin ZH, Shi X, Zheng S, Li Y, Auger C, Oikawa S, Yook JS, Granath-Panelo M, He W, Zhang GF, Matsushita M, Saito M, Gerszten RE, Mills EL, Banks AS, Ishihama Y, White PJ, McGarrah RW, Yoneshiro T, Kajimura S. BCAA-nitrogen flux in brown fat controls metabolic health independent of thermogenesis. Cell 2024; 187:2359-2374.e18. [PMID: 38653240 PMCID: PMC11145561 DOI: 10.1016/j.cell.2024.03.030] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 01/07/2024] [Accepted: 03/21/2024] [Indexed: 04/25/2024]
Abstract
Brown adipose tissue (BAT) is best known for thermogenesis. Rodent studies demonstrated that enhanced BAT thermogenesis is tightly associated with increased energy expenditure, reduced body weight, and improved glucose homeostasis. However, human BAT is protective against type 2 diabetes, independent of body weight. The mechanism underlying this dissociation remains unclear. Here, we report that impaired mitochondrial catabolism of branched-chain amino acids (BCAAs) in BAT, by deleting mitochondrial BCAA carriers (MBCs), caused systemic insulin resistance without affecting energy expenditure and body weight. Brown adipocytes catabolized BCAA in the mitochondria as nitrogen donors for the biosynthesis of non-essential amino acids and glutathione. Impaired mitochondrial BCAA-nitrogen flux in BAT resulted in increased oxidative stress, decreased hepatic insulin signaling, and decreased circulating BCAA-derived metabolites. A high-fat diet attenuated BCAA-nitrogen flux and metabolite synthesis in BAT, whereas cold-activated BAT enhanced the synthesis. This work uncovers a metabolite-mediated pathway through which BAT controls metabolic health beyond thermogenesis.
Collapse
Affiliation(s)
- Anthony R P Verkerke
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, and Howard Hughes Medical Institute, Boston, MA, USA
| | - Dandan Wang
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, and Howard Hughes Medical Institute, Boston, MA, USA
| | - Naofumi Yoshida
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, and Howard Hughes Medical Institute, Boston, MA, USA
| | - Zachary H Taxin
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, and Howard Hughes Medical Institute, Boston, MA, USA
| | - Xu Shi
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Shuning Zheng
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Yuka Li
- Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Christopher Auger
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, and Howard Hughes Medical Institute, Boston, MA, USA
| | - Satoshi Oikawa
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, and Howard Hughes Medical Institute, Boston, MA, USA
| | - Jin-Seon Yook
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, and Howard Hughes Medical Institute, Boston, MA, USA
| | - Melia Granath-Panelo
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, and Howard Hughes Medical Institute, Boston, MA, USA
| | - Wentao He
- Duke Molecular Physiology Institute, Duke School of Medicine, Department of Medicine, Division of Endocrinology, Metabolism and Nutrition, Duke University, Durham, NC, USA
| | - Guo-Fang Zhang
- Duke Molecular Physiology Institute, Duke School of Medicine, Department of Medicine, Division of Endocrinology, Metabolism and Nutrition, Duke University, Durham, NC, USA
| | - Mami Matsushita
- Department of Nutrition, School of Nursing and Nutrition, Tenshi College, Sapporo, Japan
| | - Masayuki Saito
- Laboratory of Biochemistry, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Robert E Gerszten
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Evanna L Mills
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute and Department of Immunology, Harvard Medical School, Boston, MA, USA
| | - Alexander S Banks
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Yasushi Ishihama
- Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Phillip J White
- Duke Molecular Physiology Institute, Duke School of Medicine, Department of Medicine, Division of Endocrinology, Metabolism and Nutrition, Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
| | - Robert W McGarrah
- Duke Molecular Physiology Institute, Duke School of Medicine, Sarah W. Stedman Nutrition and Metabolism Center, Department of Medicine, Division of Cardiology, Duke University, Durham, NC, USA
| | - Takeshi Yoneshiro
- Division of Metabolic Medicine, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan; Division of Molecular Physiology and Metabolism, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Shingo Kajimura
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, and Howard Hughes Medical Institute, Boston, MA, USA.
| |
Collapse
|
31
|
Figueiredo JC, Bhowmick NA, Karlstaedt A. Metabolic basis of cardiac dysfunction in cancer patients. Curr Opin Cardiol 2024; 39:138-147. [PMID: 38386340 PMCID: PMC11185275 DOI: 10.1097/hco.0000000000001118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/23/2024]
Abstract
PURPOSE OF REVIEW The relationship between metabolism and cardiovascular diseases is complex and bidirectional. Cardiac cells must adapt metabolic pathways to meet biosynthetic demands and energy requirements to maintain contractile function. During cancer, this homeostasis is challenged by the increased metabolic demands of proliferating cancer cells. RECENT FINDINGS Tumors have a systemic metabolic impact that extends beyond the tumor microenvironment. Lipid metabolism is critical to cancer cell proliferation, metabolic adaptation, and increased cardiovascular risk. Metabolites serve as signals which provide insights for diagnosis and prognosis in cardio-oncology patients. SUMMARY Metabolic processes demonstrate a complex relationship between cancer cell states and cardiovascular remodeling with potential for therapeutic interventions.
Collapse
Affiliation(s)
- Jane C. Figueiredo
- Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Neil Adri Bhowmick
- Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA
- Division of Hematology and Oncology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Anja Karlstaedt
- Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA
| |
Collapse
|
32
|
Vadadokhau U, Varga I, Káplár M, Emri M, Csősz É. Examination of the Complex Molecular Landscape in Obesity and Type 2 Diabetes. Int J Mol Sci 2024; 25:4781. [PMID: 38732002 PMCID: PMC11084226 DOI: 10.3390/ijms25094781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 04/10/2024] [Accepted: 04/25/2024] [Indexed: 05/13/2024] Open
Abstract
The escalating prevalence of metabolic disorders, notably type 2 diabetes (T2D) and obesity, presents a critical global health challenge, necessitating deeper insights into their molecular underpinnings. Our study integrates proteomics and metabolomics analyses to delineate the complex molecular landscapes associated with T2D and obesity. Leveraging data from 130 subjects, including individuals with T2D and obesity as well as healthy controls, we elucidate distinct molecular signatures and identify novel biomarkers indicative of disease progression. Our comprehensive characterization of cardiometabolic proteins and serum metabolites unveils intricate networks of biomolecular interactions and highlights differential protein expression patterns between T2D and obesity cohorts. Pathway enrichment analyses reveal unique mechanisms underlying disease development and progression, while correlation analyses elucidate the interplay between proteomics, metabolomics, and clinical parameters. Furthermore, network analyses underscore the interconnectedness of cardiometabolic proteins and provide insights into their roles in disease pathogenesis. Our findings may help to refine diagnostic strategies and inform the development of personalized interventions, heralding a new era in precision medicine and healthcare innovation. Through the integration of multi-omics approaches and advanced analytics, our study offers a crucial framework for deciphering the intricate molecular underpinnings of metabolic disorders and paving the way for transformative therapeutic strategies.
Collapse
Affiliation(s)
- Uladzislau Vadadokhau
- Proteomics Core Facility, Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
- Doctoral School of Molecular Cellular and Immune Biology, University of Debrecen, 4032 Debrecen, Hungary
| | - Imre Varga
- Department of IT Systems and Networks, Faculty of Informatics, University of Debrecen, 4028 Debrecen, Hungary;
| | - Miklós Káplár
- Department of Internal Medicine, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary;
| | - Miklós Emri
- Department of Medical Imaging, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary;
| | - Éva Csősz
- Proteomics Core Facility, Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| |
Collapse
|
33
|
Yousf S, Batra HS, Jha RM, Sardesai DM, Ananthamohan K, Chugh J, Sharma S. Identification of potential serum biomarkers associated with HbA1c levels in Indian type 2 diabetic subjects using NMR-based metabolomics. Clin Chim Acta 2024; 557:117857. [PMID: 38484908 DOI: 10.1016/j.cca.2024.117857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 02/29/2024] [Accepted: 03/02/2024] [Indexed: 03/25/2024]
Abstract
BACKGROUND The prevalence of type 2 diabetes mellitus (T2DM), a progressive metabolic disorder characterized by chronic hyperglycemia and the development of insulin resistance, has increased globally, with worrying statistics coming from children, adolescents, and young adults from developing countries like India. Here, we investigated unique circulating metabolic signatures associated with prediabetes and T2DM in an Indian cohort using NMR-based metabolomics. MATERIALS AND METHODS The study subjects included healthy volunteers (N = 101), prediabetic subjects (N = 75), and T2DM patients (N = 108). Serum metabolic profiling was performed using 1H NMR spectroscopy and major perturbed metabolites were identified by multivariate analysis and receiver operating characteristic (ROC) modules. RESULTS Of the 36 aqueous abundant metabolites, 24 showed a statistically significant difference between healthy volunteers, prediabetics, and established T2DM subjects. On performing multivariate ROC curve analysis with 5 commonly dysregulated metabolites (namely, glucose, pyroglutamate, o-phosphocholine, serine, and methionine) in prediabetes and T2DM, AUC values obtained were 0.96 (95 % confidence interval (CI) = 0.93, 0.98) for T2DM; and 0.88 (95 % CI = 0.81, 0.93) for prediabetic subjects, respectively. CONCLUSION We propose that the identified metabolite panel can be used in the future as a biomarker for clinical diagnosis, patient surveillance, and for predicting individuals at risk for developing diabetes.
Collapse
Affiliation(s)
- Saleem Yousf
- Department of Chemistry, Indian Institute of Science Education and Research (IISER), Dr. Homi Bhabha Road, Pashan, Pune 411008, India; Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Hitender S Batra
- Department of Biochemistry, Armed Forces Medical College (AFMC), Wanowrie, Pune 411040, India; Department of Biochemistry, Symbiosis Medical College for Women, Pune 412115, India.
| | - Rakesh M Jha
- Department of Biotechnology, Savitribai Phule Pune University, Ganeshkhind Road, Pune 411007, India
| | - Devika M Sardesai
- Department of Biotechnology, Savitribai Phule Pune University, Ganeshkhind Road, Pune 411007, India
| | - Kalyani Ananthamohan
- Department of Biotechnology, Savitribai Phule Pune University, Ganeshkhind Road, Pune 411007, India
| | - Jeetender Chugh
- Department of Chemistry, Indian Institute of Science Education and Research (IISER), Dr. Homi Bhabha Road, Pashan, Pune 411008, India.
| | - Shilpy Sharma
- Department of Biotechnology, Savitribai Phule Pune University, Ganeshkhind Road, Pune 411007, India.
| |
Collapse
|
34
|
Perry AS, Piaggi P, Huang S, Nayor M, Freedman J, North K, Below J, Clish C, Murthy VL, Krakoff J, Shah RV. Human metabolic chambers reveal a coordinated metabolic-physiologic response to nutrition. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.04.08.24305087. [PMID: 38645000 PMCID: PMC11030300 DOI: 10.1101/2024.04.08.24305087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
The emerging field of precision nutrition is based on the notion that inter-individual responses across diets of different calorie-macronutrient content may contribute to inter-individual differences in metabolism, adiposity, and weight gain. Free-living diet studies have been traditionally challenged by difficulties in controlling adherence to prescribed calories and macronutrient content and rarely allow a period of metabolic stability prior to metabolic measures (to minimize influences of weight changes). In this context, key physiologic measures central to precision nutrition responses may be most precisely quantified via whole room indirect calorimetry over 24-h, in which precise control of activity and nutrition can be achieved. In addition, these studies represent unique "N of 1" human crossover metabolic-physiologic experiments during which specific molecular pathways central to nutrient metabolism may be discerned. Here, we quantified 263 circulating metabolites during a ≈40-day inpatient admission in which up to 94 participants underwent seven monitored 24-h nutritional interventions of differing macronutrient composition in a whole-room indirect calorimeter to capture precision metabolic responses. Broadly, we observed heterogenous responses in metabolites across dietary chambers, with the exception of carnitines which tracked with 24-h respiratory quotient. We identified excursions in shared metabolic species (e.g., carnitines, glycerophospholipids, amino acids) that mapped onto gold-standard calorimetric measures of substrate oxidation preference and lipid availability. These findings support a coordinated metabolic-physiologic response to nutrition, highlighting the relevance of these controlled settings to uncover biological pathways of energy utilization during precision nutrition studies.
Collapse
|
35
|
Gao WY, Tian MY, Li ML, Gao SR, Wei XL, Gao C, Zhou YY, Li T, Wang HJ, Bian BL, Si N, Zhao W, Zhao HY. Study on the potential mechanism of Qingxin Lianzi Yin Decoction on renoprotection in db/db mice via network pharmacology and metabolomics. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 126:155222. [PMID: 38382279 DOI: 10.1016/j.phymed.2023.155222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 11/04/2023] [Accepted: 11/13/2023] [Indexed: 02/23/2024]
Abstract
BACKGROUND Diabetic nephropathy (DN) was one of the most popular and most significant microvascular complications of diabetes mellitus. Qingxin Lianzi Yin Decoction (QXLZY) was a traditional Chinese classical formula, suitable for chronic urinary system diseases. QXLZY had good clinical efficacy in early DN, but the underlying molecular mechanism remained unrevealed. PURPOSE This study aimed to establish the content determination method of QXLZY index components and explore the mechanism of QXLZY on DN by network pharmacology and metabolomics studies. METHODS Firstly, the content determination methods of QXLZY were established with calycosin-7-O-β-d-glucoside, acteoside, baicalin and glycyrrhizic acid as index components. Secondly, pharmacological experiments of QXLZY were evaluated using db/db mice. UHPLC-LTQ-Orbitrap MS was used to carry out untargeted urine metabolomics, serum metabolomics, and kidney metabolomics studies. Thirdly, employing network pharmacology, key components and targets were analyzed. Finally, targeted metabolomics studies were performed on the endogenous constituents in biological samples for validation based on untargeted metabolomics results. RESULTS A method for the simultaneous determination of multiple index components in QXLZY was established, which passed the comprehensive methodological verification. It was simple, feasible, and scientific. The QXLZY treatment alleviated kidney injury of db/db mice, included the degree of histopathological damage and the level of urinary microalbumin/creatinine ratio. Untargeted metabolomics studies had identified metabolic dysfunction in pathways associated with amino acid metabolism in db/db mice. Treatment with QXLZY could reverse metabolite abnormalities and influence the pathways related to energy metabolism and amino acid metabolism. It had been found that pathways with a high degree were involved in signal transduction, prominently on amino acids metabolism and lipid metabolism, analyzed by network pharmacology. Disorders of amino acid metabolism did occur in db/db mice. QXLZY could revert the levels of metabolites, such as quinolinic acid, arginine, and asparagine. CONCLUSION This study was the first time to demonstrate that QXLZY alleviated diabetes-induced pathological changes in the kidneys of db/db mice by correcting disturbances in amino acid metabolism. This work could provide a new experimental basis and theoretical guidance for the rational application of QXLZY on DN, exploring the new pharmacological effect of traditional Chinese medicine, and promoting in-depth research and development.
Collapse
Affiliation(s)
- Wen-Ya Gao
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Meng-Yao Tian
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Ming-Li Li
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Shuang-Rong Gao
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Xiao-Lu Wei
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Chang Gao
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Yan-Yan Zhou
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Tao Li
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China; Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Hong-Jie Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Bao-Lin Bian
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Nan Si
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| | - Wei Zhao
- Center for Drug Evaluation, National Medical Products Administration, Beijing 100022, China.
| | - Hai-Yu Zhao
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| |
Collapse
|
36
|
Xing X, Sun Q, Wang R, Wang Y, Wang R. Impacts of glutamate, an exercise-responsive metabolite on insulin signaling. Life Sci 2024; 341:122471. [PMID: 38301875 DOI: 10.1016/j.lfs.2024.122471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 01/24/2024] [Accepted: 01/25/2024] [Indexed: 02/03/2024]
Abstract
AIMS Disruption of the insulin signaling pathway leads to insulin resistance (IR). IR is characterized by impaired glucose and lipid metabolism. Elevated levels of circulating glutamate are correlated with metabolic indicators and may potentially predict the onset of metabolic diseases. Glutamate receptor antagonists have significantly enhanced insulin sensitivity, and improved glucose and lipid metabolism. Exercise is a well-known strategy to combat IR. The aims of our narrative review are to summarize preclinical and clinical findings to show the correlations between circulating glutamate levels, IR and metabolic diseases, discuss the causal role of excessive glutamate in IR and metabolic disturbance, and present an overview of the exercise-induced alteration in circulating glutamate levels. MATERIALS AND METHODS A literature search was conducted to identify studies on glutamate, insulin signaling, and exercise in the PubMed database. The search covered articles published from December 1955 to January 2024, using the search terms of "glutamate", "glutamic acid", "insulin signaling", "insulin resistance", "insulin sensitivity", "exercise", and "physical activity". KEY FINDINGS Elevated levels of circulating glutamate are correlated with IR. Excessive glutamate can potentially hinder the insulin signaling pathway through various mechanisms, including the activation of ectopic lipid accumulation, inflammation, and endoplasmic reticulum stress. Glutamate can also modify mitochondrial function through Ca2+ and induce purine degradation mediated by AMP deaminase 2. Exercise has the potential to decrease circulating levels of glutamate, which can be attributed to accelerated glutamate catabolism and enhanced glutamate uptake. SIGNIFICANCE Glutamate may act as a mediator in the exercise-induced improvement of insulin sensitivity.
Collapse
Affiliation(s)
- Xiaorui Xing
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China
| | - Qin Sun
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China
| | - Ruwen Wang
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China
| | - Yibing Wang
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China.
| | - Ru Wang
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China.
| |
Collapse
|
37
|
Zhang Y, Huang Z, Han W, Wu J, Li S, Qin T, Zhang C, Shi M, Han S, Gao B, Jin S, Xiao Y, Xu K, Ye W. Glutamine suppresses senescence and promotes autophagy through glycolysis inhibition-mediated AMPKα lactylation in intervertebral disc degeneration. Commun Biol 2024; 7:325. [PMID: 38486093 PMCID: PMC10940657 DOI: 10.1038/s42003-024-06000-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 03/01/2024] [Indexed: 03/18/2024] Open
Abstract
Regulating metabolic disorders has become a promising focus in treating intervertebral disc degeneration (IDD). A few drugs regulating metabolism, such as atorvastatin, metformin, and melatonin, show positive effects in treating IDD. Glutamine participates in multiple metabolic processes, including glutaminolysis and glycolysis; however, its impact on IDD is unclear. The current study reveals that glutamine levels are decreased in severely degenerated human nucleus pulposus (NP) tissues and aging Sprague-Dawley (SD) rat nucleus pulposus tissues, while lactate accumulation and lactylation are increased. Supplementary glutamine suppresses glycolysis and reduces lactate production, which downregulates adenosine-5'-monophosphate-activated protein kinase α (AMPKα) lactylation and upregulates AMPKα phosphorylation. Moreover, glutamine treatment reduces NP cell senescence and enhances autophagy and matrix synthesis via inhibition of glycolysis and AMPK lactylation, and glycolysis inhibition suppresses lactylation. Our results indicate that glutamine could prevent IDD by glycolysis inhibition-decreased AMPKα lactylation, which promotes autophagy and suppresses NP cell senescence.
Collapse
Affiliation(s)
- Yangyang Zhang
- Department of Spine Surgery, the First Affiliated Hospital of University of South China, Hengyang, 421200, China
- Department of Spine Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, 510289, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510289, China
| | - Zhengqi Huang
- Department of Spine Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, 510289, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510289, China
| | - Weitao Han
- Department of Spine Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, 510289, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510289, China
| | - Jiajun Wu
- Department of Spine Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, 510289, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510289, China
| | - Shuangxing Li
- Department of Spine Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, 510289, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510289, China
| | - Tianyu Qin
- Department of Spine Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, 510289, China
- Department of Orthopedics, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518031, China
| | - Chao Zhang
- Department of Spine Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, 510289, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510289, China
| | - Ming Shi
- Department of Spine Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, 510289, China
- Department of Orthopedics, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518031, China
| | - Shun Han
- Department of Spine Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, 510289, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510289, China
| | - Bo Gao
- Department of Spine Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, 510289, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510289, China
| | - Song Jin
- Department of Orthopedics, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518031, China
| | - Yin Xiao
- School of Medicine and Dentistry, Menzies Health Institute Queensland, Griffith University, Brisbane, QLD, Australia
| | - Kang Xu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510289, China.
| | - Wei Ye
- Department of Spine Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, 510289, China.
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510289, China.
| |
Collapse
|
38
|
Abdi F, Mohammadzadeh M, Abbasalizad-Farhangi M. Dietary amino acid patterns and cardiometabolic risk factors among subjects with obesity; a cross-sectional study. BMC Endocr Disord 2024; 24:21. [PMID: 38355488 PMCID: PMC10865612 DOI: 10.1186/s12902-024-01549-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 01/29/2024] [Indexed: 02/16/2024] Open
Abstract
BACKGROUND The prevalence of obesity is a growing global public health concern. Certain dietary amino acids have been shown to have a potential therapeutic role in improving metabolic syndrome parameters and body composition in individuals with obesity. However, some amino acids have been linked to an increased risk of cardiometabolic disorders. This cross-sectional study aims to investigate the association between dietary amino acid patterns and cardiometabolic risk factors in individuals with obesity. METHODS This cross-sectional study included 335 participants with obesity (57.9% males and 41.5% females) from Tabriz and Tehran, Iran. The participants were between the ages of 20-50, with a body mass index (BMI) of 30 kg/m2 or higher, and free from certain medical conditions. The study examined participants' general characteristics, conducted anthropometric assessments, dietary assessments, and biochemical assessments. The study also used principal component analysis to identify amino acid intake patterns and determined the association between these patterns and cardiometabolic risk factors in individuals with obesity. RESULTS Upon adjusting for potential confounders, the study found that individuals in the third tertiles of pattern 1 and 2 were more likely to have lower LDL levels (OR = 0.99 and 95% CI (0.98-0.99)) for both. Additionally, a significant decrease in total cholesterol was observed in the third tertiles of pattern 2 in model II (OR = 0.99, 95% CI (0.98-0.99)). These findings suggest a potential cardioprotective effect of these amino acid patterns in managing cardiometabolic risk factors in individuals with obesity. CONCLUSIONS This study found that two identified amino acid patterns were associated with lower serum LDL and total cholesterol levels, while a third pattern was associated with higher serum triglycerides. The specific amino acids contributing to these patterns highlight the importance of targeted dietary interventions in managing cardiometabolic risk factors in individuals with obesity.
Collapse
Affiliation(s)
- Fatemeh Abdi
- Department of Community Nutrition, Faculty of Nutrition, Tabriz University of Medical Sciences, Attar Neyshabouri, Daneshgah Blv, Tabriz, Iran
| | - Milad Mohammadzadeh
- Department of Clinical Nutrition and Dietetics, Faculty of Nutrition Sciences and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahdieh Abbasalizad-Farhangi
- Department of Community Nutrition, Faculty of Nutrition, Tabriz University of Medical Sciences, Attar Neyshabouri, Daneshgah Blv, Tabriz, Iran.
| |
Collapse
|
39
|
Thaker VV, Kwee LC, Chen H, Bahson J, Ilkayeva O, Muehlbauer MJ, Wolfe B, Purnell JQ, Pi-Sunyer X, Newgard CB, Shah SH, Laferrère B, Look AHEAD Research Group. Metabolite signature of diabetes remission in individuals with obesity undergoing weight loss interventions. Obesity (Silver Spring) 2024; 32:304-314. [PMID: 37962326 PMCID: PMC11201087 DOI: 10.1002/oby.23943] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 08/25/2023] [Accepted: 09/19/2023] [Indexed: 11/15/2023]
Abstract
OBJECTIVE This observational study investigated metabolomic changes in individuals with type 2 diabetes (T2D) after weight loss. We hypothesized that metabolite changes associated with T2D-relevant phenotypes are signatures of improved health. METHODS Fasting plasma samples from individuals undergoing bariatric surgery (n = 71 Roux-en-Y gastric bypass [RYGB], n = 22 gastric banding), lifestyle intervention (n = 66), or usual care (n = 14) were profiled for 139 metabolites before and 2 years after weight loss. Principal component analysis grouped correlated metabolites into factors. Association of preintervention metabolites was tested with preintervention clinical features and changes in T2D markers. Association between change in metabolites/metabolite factors and change in T2D remission markers, homeostasis model assessment of β-cell function, homeostasis model assessment of insulin resistance, and glycated hemoglobin (HbA1c) was assessed. RESULTS Branched-chain amino acids (BCAAs) were associated with preintervention adiposity. Changes in BCAAs (valine, leucine/isoleucine) and branched-chain ketoacids were positively associated with change in HbA1c (false discovery rate q value ≤ 0.001) that persisted after adjustment for percentage weight change and RYGB (p ≤ 0.02). In analyses stratified by RYGB or other weight loss method, some metabolites showed association with non-RYGB weight loss. CONCLUSIONS This study confirmed known metabolite associations with obesity/T2D and showed an association of BCAAs with HbA1c change after weight loss, independent of the method or magnitude of weight loss.
Collapse
Affiliation(s)
- Vidhu V. Thaker
- Department of Pediatrics, Columbia University Irving Medical Center, New York, NY
| | | | - Haiying Chen
- Department of Biostatistics and Data Science, Wake Forest School of Medicine, Salem, NC
| | - Judy Bahson
- Department of Biostatistics and Data Science, Wake Forest School of Medicine, Salem, NC
| | - Olga Ilkayeva
- Duke Molecular Physiology Institute
- Sarah W. Stedman Nutrition and Metabolism Center
- Department of Medicine, Division of Endocrinology, Metabolism, and Nutrition, Duke University School of Medicine, Durham, NC
| | | | - Bruce Wolfe
- Departments of Surgery and Medicine, Oregon Health & Science University, Portland, OR
| | - Jonathan Q Purnell
- Departments of Surgery and Medicine, Oregon Health & Science University, Portland, OR
| | - Xavier Pi-Sunyer
- New York Obesity Research Center, Division of Endocrinology, Department of Medicine, Columbia University Irving Medical Center, New York, NY
| | - Christopher B. Newgard
- Duke Molecular Physiology Institute
- Sarah W. Stedman Nutrition and Metabolism Center
- Department of Pharmacology & Cancer Biology and Division of Endocrinology, Department of Medicine, Duke University, Durham, NC
| | - Svati H. Shah
- Duke Molecular Physiology Institute
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, NC
| | - Blandine Laferrère
- New York Obesity Research Center, Division of Endocrinology, Department of Medicine, Columbia University Irving Medical Center, New York, NY
| | | |
Collapse
|
40
|
Alqudah A, Qnais E, Wedyan M, Awali A, Bseiso Y, Gammoh O. Amino acid profiles: exploring their diagnostic and pathophysiological significance in hypertension. Mol Biol Rep 2024; 51:200. [PMID: 38270677 DOI: 10.1007/s11033-023-09107-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 12/05/2023] [Indexed: 01/26/2024]
Abstract
Hypertension, a major contributor to cardiovascular morbidity, is closely linked to amino acid metabolism. Amino acids, particularly branched-chain amino acids (BCAAs) and aromatic amino acids (AAAs), may play pivotal roles in the pathogenesis and potential management of hypertension. This review investigated the relationships between amino acid profiles, specifically BCAAs and AAAs, and hypertension, and examined their potential as diagnostic and therapeutic targets. An in-depth analysis was conducted on studies highlighting the associations of specific amino acids such as arginine, glycine, proline, glutamine, and the BCAAs and AAAs with hypertension. BCAAs and AAAs, alongside other amino acids like arginine, glycine, and proline, showed significant correlations with hypertension. These amino acids influence multiple pathways including nitric oxide synthesis, vascular remodeling, and neurotransmitter production, among others. Distinct amino acid profiles were discerned between hypertensive and non-hypertensive individuals. Amino acid profiling, particularly the levels of BCAAs and AAAs, offers promising avenues in the diagnostic and therapeutic strategies for hypertension. Future studies are crucial to confirm these findings and to delineate amino acid-based interventions for hypertension treatment.
Collapse
Affiliation(s)
- Abdelrahim Alqudah
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmaceutical Sciences, The Hashemite University, Zarqa, Jordan.
| | - Esam Qnais
- Department of Biology and Biotechnology, Faculty of Science, The Hashemite University, Zarqa, Jordan
| | - Mohammed Wedyan
- Department of Biology and Biotechnology, Faculty of Science, The Hashemite University, Zarqa, Jordan
| | - Ayat Awali
- Department of Biology and Biotechnology, Faculty of Science, The Hashemite University, Zarqa, Jordan
| | - Yousra Bseiso
- Department of Biology and Biotechnology, Faculty of Science, The Hashemite University, Zarqa, Jordan
| | - Omar Gammoh
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmacy, Yarmouk University, Irbid, Jordan
| |
Collapse
|
41
|
Islam SJ, Liu C, Mohandas AN, Rooney K, Nayak A, Mehta A, Ko YA, Kim JH, Sun YV, Dunbar SB, Lewis TT, Taylor HA, Uppal K, Jones DP, Quyyumi AA, Searles CD. Metabolomic signatures of ideal cardiovascular health in black adults. Sci Rep 2024; 14:1794. [PMID: 38245568 PMCID: PMC10799852 DOI: 10.1038/s41598-024-51920-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 01/11/2024] [Indexed: 01/22/2024] Open
Abstract
Plasma metabolomics profiling is an emerging methodology to identify metabolic pathways underlying cardiovascular health (CVH). The objective of this study was to define metabolomic profiles underlying CVH in a cohort of Black adults, a population that is understudied but suffers from disparate levels of CVD risk factors. The Morehouse-Emory Cardiovascular (MECA) Center for Health Equity study cohort consisted of 375 Black adults (age 53 ± 10, 39% male) without known CVD. CVH was determined by the AHA Life's Simple 7 (LS7) score, calculated from measured blood pressure, body mass index (BMI), fasting blood glucose and total cholesterol, and self-reported physical activity, diet, and smoking. Plasma metabolites were assessed using untargeted high-resolution metabolomics profiling. A metabolome wide association study (MWAS) identified metabolites associated with LS7 score after adjusting for age and sex. Using Mummichog software, metabolic pathways that were significantly enriched in metabolites associated with LS7 score were identified. Metabolites representative of these pathways were compared across clinical domains of LS7 score and then developed into a metabolomics risk score for prediction of CVH. We identified novel metabolomic signatures and pathways associated with CVH in a cohort of Black adults without known CVD. Representative and highly prevalent metabolites from these pathways included glutamine, glutamate, urate, tyrosine and alanine, the concentrations of which varied with BMI, fasting glucose, and blood pressure levels. When assessed in conjunction, these metabolites were independent predictors of CVH. One SD increase in the novel metabolomics risk score was associated with a 0.88 higher LS7 score, which translates to a 10.4% lower incident CVD risk. We identified novel metabolomic signatures of ideal CVH in a cohort of Black Americans, showing that a core group of metabolites central to nitrogen balance, bioenergetics, gluconeogenesis, and nucleotide synthesis were associated with CVH in this population.
Collapse
Affiliation(s)
- Shabatun J Islam
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Chang Liu
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Appesh N Mohandas
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Kimberly Rooney
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Aditi Nayak
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Anurag Mehta
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Yi-An Ko
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Jeong Hwan Kim
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Yan V Sun
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
- Department of Biomedical Informatics, Emory University School of Medicine, Atlanta, GA, USA
- Atlanta VA Health Care System, Decatur, GA, USA
| | - Sandra B Dunbar
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
- Nell Hodgson Woodruff School of Nursing, Emory University, Atlanta, GA, USA
| | - Tené T Lewis
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Herman A Taylor
- Department of Medicine, Morehouse School of Medicine, Atlanta, GA, USA
| | - Karan Uppal
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Dean P Jones
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Arshed A Quyyumi
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Charles D Searles
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA.
- Atlanta VA Health Care System, Decatur, GA, USA.
| |
Collapse
|
42
|
Yang TN, Wang YX, Jian PA, Ma XY, Zhu SY, Li XN, Li JL. Exogenous Melatonin Alleviates Atrazine-Induced Glucose Metabolism Disorders in Mice Liver via Suppressing Endoplasmic Reticulum Stress. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:742-751. [PMID: 38111124 DOI: 10.1021/acs.jafc.3c06441] [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: 12/20/2023]
Abstract
Atrazine (ATZ) is a widely used herbicide that has toxic effects on animals. Melatonin (MLT) is a natural hormone with strong antioxidant properties. However, the effect of MLT on the glucose metabolism disorder caused by ATZ is still unclear. Mice were divided into four groups randomly and given 21 days of gavage: blank control group (Con), 5 mg/kg MLT group (MLT), 170 mg/kg ATZ group (ATZ), and 170 mg/kg ATZ and 5 mg/kg MLT group (ATZ + MLT). The results show that ATZ alters mRNA levels of metabolic enzymes related to glycogen synthesis and glycolysis and increased metabolites (glycogen, lactate, and pyruvate). ATZ causes abnormalities in glucose metabolism in mouse liver, interfering with glycemia regulation ability. MLT can regulate the endoplasmic reticulum to respond to disordered glucose metabolism in mice liver. This study suggested that MLT has the power to alleviate the ATZ-induced glycogen overdeposition and glycolytic deficit.
Collapse
Affiliation(s)
- Tian-Ning Yang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P. R. China
| | - Yu-Xiang Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P. R. China
| | - Ping-An Jian
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P. R. China
| | - Xiang-Yu Ma
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P. R. China
| | - Shi-Yong Zhu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P. R. China
| | - Xue-Nan Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P. R. China
- Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Northeast Agricultural University, Harbin 150030, P. R. China
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Northeast Agricultural University, Harbin 150030, P. R. China
| | - Jin-Long Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P. R. China
- Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Northeast Agricultural University, Harbin 150030, P. R. China
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Northeast Agricultural University, Harbin 150030, P. R. China
| |
Collapse
|
43
|
Wunderle C, von Arx D, Mueller SC, Bernasconi L, Neyer P, Tribolet P, Stanga Z, Mueller B, Schuetz P. Association of Glutamine and Glutamate Metabolism with Mortality among Patients at Nutritional Risk-A Secondary Analysis of the Randomized Clinical Trial EFFORT. Nutrients 2024; 16:222. [PMID: 38257115 PMCID: PMC10821126 DOI: 10.3390/nu16020222] [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: 12/07/2023] [Revised: 01/02/2024] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
Glutamine and its metabolite glutamate serve as the main energy substrates for immune cells, and their plasma levels drop during severe illness. Therefore, glutamine supplementation in the critical care setting has been advocated. However, little is known about glutamine metabolism in severely but not critically ill medical patients. We investigated the prognostic impact of glutamine metabolism in a secondary analysis of the Effect of Early Nutritional Support on Frailty, Functional Outcomes, and Recovery of Malnourished Medical Inpatients Trial (EFFORT), a randomized controlled trial comparing individualized nutritional support to usual care in patients at nutritional risk. Among 234 patients with available measurements, low plasma levels of glutamate were independently associated with 30-day mortality (adjusted HR 2.35 [95% CI 1.18-4.67, p = 0.015]). The impact on mortality remained consistent long-term for up to 5 years. No significant association was found for circulating glutamine levels and short- or long-term mortality. There was no association of glutamate nor glutamine with malnutrition parameters or with the effectiveness of nutritional support. This secondary analysis found glutamate to be independently prognostic among medical inpatients at nutritional risk but poorly associated with the effectiveness of nutritional support. In contrast to ICU studies, we found no association between glutamine and clinical outcome.
Collapse
Affiliation(s)
- Carla Wunderle
- Division of General Internal and Emergency Medicine, University Department of Medicine, Kantonsspital Aarau, Tellstrasse 25, 5001 Aarau, Switzerland; (C.W.); (B.M.)
| | - Diana von Arx
- Medical Faculty, University of Basel, 4056 Basel, Switzerland
| | - Sydney Chiara Mueller
- Medical Faculty, University of Basel, 4056 Basel, Switzerland
- Faculty of Biomedical Scienes, Università della Svizzera italiana (USI), 6900 Lugano, Switzerland
| | - Luca Bernasconi
- Institute of Laboratory Medicine, Kantonsspital Aarau, Tellstrasse 25, 5001 Aarau, Switzerland (P.N.)
| | - Peter Neyer
- Institute of Laboratory Medicine, Kantonsspital Aarau, Tellstrasse 25, 5001 Aarau, Switzerland (P.N.)
| | - Pascal Tribolet
- Division of General Internal and Emergency Medicine, University Department of Medicine, Kantonsspital Aarau, Tellstrasse 25, 5001 Aarau, Switzerland; (C.W.); (B.M.)
- Department of Health Professions, Bern University of Applied Sciences, Murtenstrasse 10, 3008 Bern, Switzerland
- Faculty of Life Sciences, University of Vienna, Djerassiplatz 1, 1030 Vienna, Austria
| | - Zeno Stanga
- Department of Diabetes, Endocrinology, Nutritional Medicine and Metabolism (UDEM), Inselspital, Bern University Hospital, University of Bern, Freiburgstrasse 15, 3010 Bern, Switzerland
| | - Beat Mueller
- Division of General Internal and Emergency Medicine, University Department of Medicine, Kantonsspital Aarau, Tellstrasse 25, 5001 Aarau, Switzerland; (C.W.); (B.M.)
- Faculty of Biomedical Scienes, Università della Svizzera italiana (USI), 6900 Lugano, Switzerland
| | - Philipp Schuetz
- Division of General Internal and Emergency Medicine, University Department of Medicine, Kantonsspital Aarau, Tellstrasse 25, 5001 Aarau, Switzerland; (C.W.); (B.M.)
- Medical Faculty, University of Basel, 4056 Basel, Switzerland
| |
Collapse
|
44
|
Gillies NA, Milan AM, Cameron-Smith D, Mumme KD, Conlon CA, von Hurst PR, Haskell-Ramsay CF, Jones B, Roy NC, Coad J, Wall CR, Beck KL. Vitamin B and One-Carbon Metabolite Profiles Show Divergent Associations with Cardiometabolic Risk Markers but not Cognitive Function in Older New Zealand Adults: A Secondary Analysis of the REACH Study. J Nutr 2023; 153:3529-3542. [PMID: 37863266 DOI: 10.1016/j.tjnut.2023.10.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 10/05/2023] [Accepted: 10/16/2023] [Indexed: 10/22/2023] Open
Abstract
BACKGROUND Vitamin B inadequacies and elevated homocysteine status have been associated with impaired cognitive and cardiometabolic health with aging. There is, however, a scarcity of research investigating integrated profiles of one-carbon (1C) metabolites in this context, including metabolites of interconnected folate, methionine, choline oxidation, and transsulfuration pathways. OBJECTIVES The study aimed to examine associations between vitamins B and 1C metabolites with cardiometabolic health and cognitive function in healthy older adults, including the interactive effects of Apolipoprotein E-ε4 status. METHODS Three hundred and thirteen healthy participants (65-74 y, 65% female) were analyzed. Vitamins B were estimated according to dietary intake (4-d food records) and biochemical status (serum folate and vitamin B12). Fasting plasma 1C metabolites were quantified by liquid chromatography with tandem mass spectrometry. Measures of cardiometabolic health included biochemical (lipid panel, blood glucose) and anthropometric markers. Cognitive function was assessed by the Computerized Mental Performance Assessment System (COMPASS) and Montreal Cognitive Assessment (MoCA). Associations were analyzed using multivariate linear (COMPASS, cardiometabolic health) and Poisson (MoCA) regression modeling. RESULTS Over 90% of participants met dietary recommendations for riboflavin and vitamins B6 and B12, but only 78% of males and 67% of females achieved adequate folate intakes. Higher serum folate and plasma betaine and glycine concentrations were associated with favorable cardiometabolic markers, whereas higher plasma choline and homocysteine concentrations were associated with greater cardiometabolic risk based on body mass index and serum lipids concentration values (P< 0.05). Vitamins B and homocysteine were not associated with cognitive performance in this cohort, though higher glycine concentrations were associated with better global cognitive performance (P = 0.017), episodic memory (P = 0.016), and spatial memory (P = 0.027) scores. Apolipoprotein E-ε4 status did not modify the relationship between vitamins B or 1C metabolites with cognitive function in linear regression analyses. CONCLUSIONS Vitamin B and 1C metabolite profiles showed divergent associations with cardiometabolic risk markers and limited associations with cognitive performance in this cohort of healthy older adults.
Collapse
Affiliation(s)
- Nicola A Gillies
- The Liggins Institute, The University of Auckland, New Zealand; The Riddet Institute, New Zealand
| | - Amber M Milan
- The Liggins Institute, The University of Auckland, New Zealand; The High-Value Nutrition National Science Challenge, New Zealand; AgResearch Ltd, Grasslands Research Centre, New Zealand
| | - David Cameron-Smith
- The Liggins Institute, The University of Auckland, New Zealand; The Riddet Institute, New Zealand; School of Agriculture, Food and Ecosystem Sciences, The University of Melbourne, Australia
| | - Karen D Mumme
- School of Sport Exercise and Nutrition, Massey University, New Zealand
| | - Cathryn A Conlon
- School of Sport Exercise and Nutrition, Massey University, New Zealand
| | | | | | - Beatrix Jones
- Department of Statistics, University of Auckland, New Zealand; The High-Value Nutrition National Science Challenge, New Zealand
| | - Nicole C Roy
- The Riddet Institute, New Zealand; The High-Value Nutrition National Science Challenge, New Zealand; Department of Human Nutrition, University of Otago, New Zealand
| | - Jane Coad
- College of Sciences, Massey University, New Zealand
| | - Clare R Wall
- Discipline of Nutrition and Dietetics, University of Auckland, Auckland, New Zealand
| | - Kathryn L Beck
- School of Sport Exercise and Nutrition, Massey University, New Zealand.
| |
Collapse
|
45
|
Huang C, Luo Y, Zeng B, Chen Y, Liu Y, Chen W, Liao X, Liu Y, Wang Y, Wang X. Branched-chain amino acids prevent obesity by inhibiting the cell cycle in an NADPH-FTO-m 6A coordinated manner. J Nutr Biochem 2023; 122:109437. [PMID: 37666478 DOI: 10.1016/j.jnutbio.2023.109437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 08/15/2023] [Accepted: 08/30/2023] [Indexed: 09/06/2023]
Abstract
Obesity has become a major health crisis in the past decades. Branched-chain amino acids (BCAA), a class of essential amino acids, exerted beneficial health effects with regard to obesity and its related metabolic dysfunction, although the underlying reason is unknown. Here, we show that BCAA supplementation alleviates high-fat diet (HFD)-induced obesity and insulin resistance in mice and inhibits adipogenesis in 3T3-L1 cells. Further, we find that BCAA prevent the mitotic clonal expansion (MCE) of preadipocytes by reducing cyclin A2 (CCNA2) and cyclin-dependent kinase 2 (CDK2) expression. Mechanistically, BCAA decrease the concentration of nicotinamide adenine dinucleotide phosphate (NADPH) in adipose tissue and 3T3-L1 cells by reducing glucose-6-phosphate dehydrogenase (G6PD) expression. The reduced NADPH attenuates the expression of fat mass and obesity-associated (FTO) protein, a well-known m6A demethylase, to increase the N6-methyladenosine (m6A) levels of Ccna2 and Cdk2 mRNA. Meanwhile, the high m6A levels of Ccna2 and Cdk2 mRNA are recognized by YTH N6-methyladenosine RNA binding protein 2 (YTHDF2), which results in mRNA decay and reduction of their protein expressions. Overall, our data demonstrate that BCAA inhibit obesity and adipogenesis by reducing CDK2 and CCNA2 expression via an NADPH-FTO-m6A coordinated manner in vivo and in vitro, which raises a new perspective on the role of m6A in the BCAA regulation of obesity and adipogenesis.
Collapse
Affiliation(s)
- Chaoqun Huang
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang province, China; Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, China; Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, China; Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou, China
| | - Yaojun Luo
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang province, China; Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, China; Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, China; Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou, China
| | - Botao Zeng
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang province, China; Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, China; Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, China; Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou, China
| | - Yushi Chen
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang province, China; Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, China; Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, China; Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou, China
| | - Youhua Liu
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang province, China; Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, China; Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, China; Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou, China
| | - Wei Chen
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang province, China; Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, China; Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, China; Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou, China
| | - Xing Liao
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang province, China; Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, China; Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, China; Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou, China
| | - Yuxi Liu
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang province, China; Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, China; Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, China; Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou, China
| | - Yizhen Wang
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang province, China; Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, China; Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, China; Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou, China
| | - Xinxia Wang
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang province, China; Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, China; Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, China; Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou, China.
| |
Collapse
|
46
|
Chang KH, Chen CM, Lin CN, Tsai SS, Lyu RK, Chu CC, Ro LS, Liao MF, Chang HS, Weng YC, Hwang JS, Kuo HC. Identification of blood metabolic biomarkers associated with diabetic distal symmetric sensorimotor polyneuropathy in patients with type 2 diabetes mellitus. J Peripher Nerv Syst 2023; 28:651-663. [PMID: 37831393 DOI: 10.1111/jns.12600] [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] [Revised: 10/06/2023] [Accepted: 10/12/2023] [Indexed: 10/14/2023]
Abstract
BACKGROUND Distal symmetric sensorimotor polyneuropathy (DSPN) is a common neurologic complication of type 2 diabetes mellitus (T2DM), but the underlying mechanisms and changes in serum metabolites remain largely undefined. This study aimed to characterize the plasma metabolite profiles of participants with T2DM using targeted metabolomics analysis and identify potential biomarkers for DSPN. METHODS A combined liquid chromatography MS/MS and direct flow injection were used to quantify plasma metabolite obtained from 63 participants with T2DM, 81 with DSPN, and 33 nondiabetic control participants. A total of 130 metabolites, including amino acids, biogenic amines, sphingomyelins (SM), phosphatidylcholines, carnitines, and hexose, were analyzed. RESULTS A total of 16 plasma metabolites and 3 cholesterol-related laboratory parameters were found to have variable importance in the projection score >1.0 and false discovery rate <5.0% between control, T2DM, and DSPN. Among these variables, five serum metabolites, including phenylalanine (AUC = 0.653), alanine (AUC = 0.630), lysine (AUC = 0.622) tryptophan (AUC = 0.620), and SM C16:0 (AUC = 0.630), are potential biomarkers (all p < .05) in distinguishing T2DM with DSPN from those without (AUC = 0.720). CONCLUSIONS In this cross-sectional study, derangement of several metabolites in the plasma was observed in T2DM with and without DSPN, and these metabolites may be potential biomarkers for predicting DSPN. Longitudinal studies are warranted.
Collapse
Affiliation(s)
- Kuo-Hsuan Chang
- Department of Neurology, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan City, Taiwan
- College of Medicine, Chang Gung University, Taoyuan City, Taiwan
| | - Chiung-Mei Chen
- Department of Neurology, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan City, Taiwan
- College of Medicine, Chang Gung University, Taoyuan City, Taiwan
| | - Chia-Ni Lin
- Department of Laboratory Medicine, Chang Gung Memorial Hospital, Taoyuan City, Taiwan
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan City, Taiwan
| | - Sung-Sheng Tsai
- College of Medicine, Chang Gung University, Taoyuan City, Taiwan
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan City, Taiwan
| | - Rong-Kuo Lyu
- Department of Neurology, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan City, Taiwan
- College of Medicine, Chang Gung University, Taoyuan City, Taiwan
| | - Chun-Che Chu
- Department of Neurology, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan City, Taiwan
- College of Medicine, Chang Gung University, Taoyuan City, Taiwan
| | - Long-Sun Ro
- Department of Neurology, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan City, Taiwan
- College of Medicine, Chang Gung University, Taoyuan City, Taiwan
| | - Ming-Feng Liao
- Department of Neurology, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan City, Taiwan
- College of Medicine, Chang Gung University, Taoyuan City, Taiwan
| | - Hong-Shiu Chang
- Department of Neurology, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan City, Taiwan
- College of Medicine, Chang Gung University, Taoyuan City, Taiwan
| | - Yi-Ching Weng
- Department of Neurology, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan City, Taiwan
- College of Medicine, Chang Gung University, Taoyuan City, Taiwan
| | - Jawl-Shan Hwang
- College of Medicine, Chang Gung University, Taoyuan City, Taiwan
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan City, Taiwan
| | - Hung-Chou Kuo
- Department of Neurology, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan City, Taiwan
- College of Medicine, Chang Gung University, Taoyuan City, Taiwan
| |
Collapse
|
47
|
Tsukagoshi-Yamaguchi A, Koshizaka M, Ishibashi R, Ishikawa K, Ishikawa T, Shoji M, Ide S, Ide K, Baba Y, Terayama R, Hattori A, Takemoto M, Ouchi Y, Maezawa Y, Yokote K. Metabolomic analysis of serum samples from a clinical study on ipragliflozin and metformin treatment in Japanese patients with type 2 diabetes: Exploring human metabolites associated with visceral fat reduction. Pharmacotherapy 2023; 43:1317-1326. [PMID: 37772313 DOI: 10.1002/phar.2884] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 09/02/2023] [Accepted: 09/05/2023] [Indexed: 09/30/2023]
Abstract
STUDY OBJECTIVE The effects of the sodium-dependent glucose transporter-2 inhibitor ipragliflozin were compared with metformin in a previous study, which revealed that ipragliflozin reduced visceral fat content by 12%; however, the underlying mechanism was unclear. Therefore, this sub-analysis aimed to compare metabolomic changes associated with ipragliflozin and metformin that may contribute to their biological effects. DESIGN A sub-analysis of a randomized controlled study. SETTING Chiba University Hospital and ten hospitals in Japan. PATIENTS Fifteen patients with type 2 diabetes in the ipragliflozin group and 15 patients with type 2 diabetes in the metformin group with matching characteristics, such as age, sex, baseline A1C, baseline visceral fat area, smoking status, and concomitant medication. INTERVENTIONS Ipragliflozin 50 mg or metformin 1000 mg daily. MEASUREMENTS The clinical data were reanalyzed, and metabolomic analysis of serum samples collected before and 24 weeks after drug administration was performed using capillary electrophoresis time-of-flight mass spectrometry. MAIN RESULTS The reduction in the mean visceral fat area after 24 weeks of treatment was significantly larger (p = 0.002) in the ipragliflozin group (-19.8%) than in the metformin group (-2.5%), as were the subcutaneous fat area and body weight. The A1C and blood glucose levels decreased in both groups. Glutamic pyruvic oxaloacetic transaminase, γ-glutamyl transferase, uric acid, and triglyceride levels decreased in the ipragliflozin group. Low-density lipoprotein cholesterol levels decreased in the metformin group. After ipragliflozin administration, N2-phenylacetylglutamine, inosine, guanosine, and 1-methyladenosine levels increased, whereas galactosamine, glucosamine, 11-aminoundecanoic acid, morpholine, and choline levels decreased. After metformin administration, metformin, hypotaurine, methionine, methyl-2-oxovaleric acid, 3-nitrotyrosine, and cyclohexylamine levels increased, whereas citrulline, octanoic acid, indole-3-acetaldehyde, and hexanoic acid levels decreased. CONCLUSIONS Metabolites that may affect visceral fat reduction were detected in the ipragliflozin group. Studies are required to further elucidate the underlying mechanisms.
Collapse
Affiliation(s)
| | - Masaya Koshizaka
- Department of Diabetes, Metabolism and Endocrinology, Chiba University Hospital, Chiba City, Japan
- Department of Endocrinology, Hematology and Gerontology, Chiba University Graduate School of Medicine, Chiba City, Japan
- Center for Preventive Medical Science, Chiba University, Chiba City, Japan
| | - Ryoichi Ishibashi
- Department of Endocrinology, Hematology and Gerontology, Chiba University Graduate School of Medicine, Chiba City, Japan
- Division of Diabetes, Endocrinology, and Metabolism, Kimitsu Chuo Hospital, Kisarazu City, Japan
| | - Ko Ishikawa
- Department of Diabetes and Endocrinology, Chiba Rosai Hospital, Ichihara City, Japan
| | - Takahiro Ishikawa
- Department of Diabetes, Metabolism and Endocrinology, Chiba University Hospital, Chiba City, Japan
- Department of General Medical Science, Chiba University Graduate School of Medicine, Chiba City, Japan
| | - Mayumi Shoji
- Department of Diabetes, Metabolism and Endocrinology, Chiba University Hospital, Chiba City, Japan
- Department of Endocrinology, Hematology and Gerontology, Chiba University Graduate School of Medicine, Chiba City, Japan
| | - Shintaro Ide
- Department of Diabetes, Metabolism and Endocrinology, Chiba University Hospital, Chiba City, Japan
- Department of Endocrinology, Hematology and Gerontology, Chiba University Graduate School of Medicine, Chiba City, Japan
| | - Kana Ide
- Department of Diabetes, Metabolism and Endocrinology, Chiba University Hospital, Chiba City, Japan
- Department of Endocrinology, Hematology and Gerontology, Chiba University Graduate School of Medicine, Chiba City, Japan
| | - Yusuke Baba
- Department of Diabetes, Metabolism and Endocrinology, Chiba University Hospital, Chiba City, Japan
- Department of Endocrinology, Hematology and Gerontology, Chiba University Graduate School of Medicine, Chiba City, Japan
| | - Ryo Terayama
- Department of Diabetes, Metabolism and Endocrinology, Chiba University Hospital, Chiba City, Japan
| | - Akiko Hattori
- Department of Diabetes, Metabolism and Endocrinology, Chiba University Hospital, Chiba City, Japan
- Department of Endocrinology, Hematology and Gerontology, Chiba University Graduate School of Medicine, Chiba City, Japan
| | - Minoru Takemoto
- Division of Diabetes, Department of Medicine, Metabolism and Endocrinology, International University of Health and Welfare, Narita City, Japan
| | - Yasuo Ouchi
- Department of Regenerative Medicine, Chiba University Graduate School of Medicine, Chiba City, Japan
- Altos Labs, California, San Diego, USA
| | - Yoshiro Maezawa
- Department of Diabetes, Metabolism and Endocrinology, Chiba University Hospital, Chiba City, Japan
- Department of Endocrinology, Hematology and Gerontology, Chiba University Graduate School of Medicine, Chiba City, Japan
| | - Koutaro Yokote
- Department of Diabetes, Metabolism and Endocrinology, Chiba University Hospital, Chiba City, Japan
- Department of Endocrinology, Hematology and Gerontology, Chiba University Graduate School of Medicine, Chiba City, Japan
| |
Collapse
|
48
|
Najafi F, Mohseni P, Pasdar Y, Niknam M, Izadi N. The association between dietary amino acid profile and the risk of type 2 diabetes: Ravansar non-communicable disease cohort study. BMC Public Health 2023; 23:2284. [PMID: 37980456 PMCID: PMC10657569 DOI: 10.1186/s12889-023-17210-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Accepted: 11/11/2023] [Indexed: 11/20/2023] Open
Abstract
BACKGROUND Type 2 diabetes (T2D) is one of the most common chronic diseases and the main risk factors for T2D consist of a combination of lifestyle, unhealthy diet, and genetic factors. Amino acids are considered to be a major component of dietary sources for many of the associations between dietary protein and chronic disease. Therefore, this study amied to determine the association between dietary amino acid intakes and the incidence of T2D. METHODS The present nested case-control study was conducted using data from the Ravansar Non-Communicable Disease (RaNCD) Cohort Study. The information required for this study was collected from individuals who participated in the Adult Cohort Study from the start of the study until September 2023. Over a 6-year follow-up period, data from 113 new T2D cases were available. Four controls were then randomly selected for each case using density sampling. Cases and controls were matched for sex and age at the interview. Food frequency questionnaire (FFQ) was used to collect data related to all amino acids including tryptophan, threonine, isoleucine, leucine, lysine, methionine, cysteine, phenylalanine, tyrosine, valine, arginine, histidine, alanine, aspartic acid, glutamic acid, glycine, proline, and serine were also extracted. Binary logistic regression was used to estimate the crude and adjusted odds ratio for the risk of T2D. RESULTS Using the univariable model, a significant association was found between T2D risk and branched-chain, alkaline, sulfuric, and essential amino acids in the fourth quartile. Accordingly, individuals in the fourth quartile had a 1.81- to 1.87-fold higher risk of developing new T2D than individuals in the lowest quartile (P<0.05). After adjustment for several variables, the risk of developing a new T2D was 2.70 (95% CI: 1.16-6.31), 2.68 (95% CI: 1.16-6.21), 2.98 (95% CI: 1.27-6.96), 2.45 (95% CI: 1.02-5.90), and 2.66 (95% CI: 1.13-6.25) times higher, for individuals in the fourth quartile of branched-chain, alkaline, sulfuric, alcoholic, and essential amino acids compared with those in the lowest quartile, respectively. CONCLUSIONS The results showed that the risk of developing a new T2D was higher for individuals in the fourth quartile of branched-chain amino acids, alkaline, sulfate, and essential amino acids than in the lower quartile.
Collapse
Affiliation(s)
- Farid Najafi
- Research Center for Environmental Determinants of Health (RCEDH), Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Parisa Mohseni
- Department of Epidemiology, School of Public Health and Safety, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Yahya Pasdar
- Research Center for Environmental Determinants of Health (RCEDH), Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mahdieh Niknam
- Research Center for Social Determinants of Health, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Neda Izadi
- Research Center for Environmental Determinants of Health (RCEDH), Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
49
|
Pan X, Ye L, Guo X, Wang W, Zhang Z, Wang Q, Huang J, Xu J, Cai Y, Shou X, Wang Y, Feng Y, Xie C, Shan P, Meng ZX. Glutamine Production by Glul Promotes Thermogenic Adipocyte Differentiation Through Prdm9-Mediated H3K4me3 and Transcriptional Reprogramming. Diabetes 2023; 72:1574-1596. [PMID: 37579296 DOI: 10.2337/db23-0162] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Accepted: 08/09/2023] [Indexed: 08/16/2023]
Abstract
Thermogenic adipocytes have been extensively investigated because of their energy-dissipating property and therapeutic potential for obesity and diabetes. Besides serving as fuel sources, accumulating evidence suggests that intermediate metabolites play critical roles in multiple biological processes. However, their role in adipocyte differentiation and thermogenesis remains unexplored. Here, we report that human and mouse obesity is associated with marked downregulation of glutamine synthetase (Glul) expression and activity in thermogenic adipose tissues. Glul is robustly upregulated during brown adipocyte (BAC) differentiation and in brown adipose tissue (BAT) upon cold exposure and Cl316,243 stimulation. Further genetic, pharmacologic, or metabolic manipulations of Glul and glutamine levels reveal that glutamine cells autonomously stimulate BAC differentiation and function and BAT remodeling and improve systemic energy homeostasis in mice. Mechanistically, glutamine promotes transcriptional induction of adipogenic and thermogenic gene programs through histone modification-mediated chromatin remodeling. Among all the glutamine-regulated writer and eraser genes responsible for histone methylation and acetylation, only Prdm9, a histone lysine methyltransferase, is robustly induced during BAC differentiation. Importantly, Prdm9 inactivation by shRNA knockdown or a selective inhibitor attenuates glutamine-triggered adipogenic and thermogenic induction. Furthermore, Prdm9 gene transcription is regulated by glutamine through the recruitment of C/EBPb to its enhancer region. This work reveals glutamine as a novel activator of thermogenic adipocyte differentiation and uncovers an unexpected role of C/EBPb-Prdm9-mediated H3K4me3 and transcriptional reprogramming in adipocyte differentiation and thermogenesis. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Xiaowen Pan
- Department of Pathology and Pathophysiology and Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Key Laboratory of Disease Proteomics of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Lingxia Ye
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xiaozhen Guo
- State Key Laboratory of Drug Research, Shanghai Institute of Material Medical, Chinese Academy of Sciences, Shanghai, China
| | - Weihua Wang
- Department of Pathology and Pathophysiology and Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Key Laboratory of Disease Proteomics of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Ziyin Zhang
- Department of Pathology and Pathophysiology and Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Key Laboratory of Disease Proteomics of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Qintao Wang
- Department of Pathology and Pathophysiology and Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Key Laboratory of Disease Proteomics of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jingjing Huang
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jingya Xu
- Department of Pathology and Pathophysiology and Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Key Laboratory of Disease Proteomics of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yanhan Cai
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xinxin Shou
- Department of Pathology and Pathophysiology and Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yuting Wang
- Department of Endocrinology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Yu Feng
- Department of Endocrinology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Cen Xie
- State Key Laboratory of Drug Research, Shanghai Institute of Material Medical, Chinese Academy of Sciences, Shanghai, China
| | - Pengfei Shan
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Zhuo-Xian Meng
- Department of Pathology and Pathophysiology and Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Key Laboratory of Disease Proteomics of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Department of Geriatrics, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Chronic Disease Research Institute, Zhejiang University School of Public Health, Hangzhou, China
| |
Collapse
|
50
|
Bishehsari F, Drees M, Adnan D, Sharma D, Green S, Koshy J, Giron LB, Goldman A, Abdel-Mohsen M, Rasmussen HE, Miller GE, Keshavarzian A. Multi-omics approach to socioeconomic disparity in metabolic syndrome reveals roles of diet and microbiome. Proteomics 2023; 23:e2300023. [PMID: 37525324 DOI: 10.1002/pmic.202300023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 06/23/2023] [Accepted: 07/10/2023] [Indexed: 08/02/2023]
Abstract
The epidemy of metabolic syndrome (MetS) is typically preceded by adoption of a "risky" lifestyle (e.g., dietary habit) among populations. Evidence shows that those with low socioeconomic status (SES) are at an increased risk for MetS. To investigate this, we recruited 123 obese subjects (body mass index [BMI] ≥ 30) from Chicago. Multi-omic data were collected to interrogate fecal microbiota, systemic markers of inflammation and immune activation, plasma metabolites, and plasma glycans. Intestinal permeability was measured using the sugar permeability testing. Our results suggest a heterogenous metabolic dysregulation among obese populations who are at risk of MetS. Systemic inflammation, linked to poor diet, intestinal microbiome dysbiosis, and gut barrier dysfunction may explain the development of MetS in these individuals. Our analysis revealed 37 key features associated with increased numbers of MetS features. These features were used to construct a composite metabolic-inflammatory (MI) score that was able to predict progression of MetS among at-risk individuals. The MI score was correlated with several markers of poor diet quality as well as lower levels of gut microbial diversity and abnormalities in several species of bacteria. This study reveals novel targets to reduce the burden of MetS and suggests access to healthy food options as a practical intervention.
Collapse
Affiliation(s)
- Faraz Bishehsari
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, Illinois, USA
| | - Michael Drees
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, Illinois, USA
| | - Darbaz Adnan
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, Illinois, USA
| | - Deepak Sharma
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, Illinois, USA
| | - Stefan Green
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, Illinois, USA
| | - Jane Koshy
- The Wistar Institute, Philadelphia, Pennsylvania, USA
| | - Leila B Giron
- The Wistar Institute, Philadelphia, Pennsylvania, USA
| | - Aaron Goldman
- The Wistar Institute, Philadelphia, Pennsylvania, USA
| | | | | | - Gregory E Miller
- Institute for Policy Research and Dept of Psychology, Northwestern Univ, Evanston, Illinois, USA
| | - Ali Keshavarzian
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, Illinois, USA
| |
Collapse
|