101
|
Francis EC, Kechris K, Cohen CC, Michelotti G, Dabelea D, Perng W. Metabolomic Profiles in Childhood and Adolescence Are Associated with Fetal Overnutrition. Metabolites 2022; 12:265. [PMID: 35323708 PMCID: PMC8952572 DOI: 10.3390/metabo12030265] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 03/07/2022] [Accepted: 03/16/2022] [Indexed: 02/01/2023] Open
Abstract
Fetal overnutrition predisposes offspring to increased metabolic risk. The current study used metabolomics to assess sustained differences in serum metabolites across childhood and adolescence among youth exposed to three typologies of fetal overnutrition: maternal obesity only, gestational diabetes mellitus (GDM) only, and obesity + GDM. We included youth exposed in utero to obesity only (BMI ≥ 30; n = 66), GDM only (n = 56), obesity + GDM (n = 25), or unexposed (n = 297), with untargeted metabolomics measured at ages 10 and 16 years. We used linear mixed models to identify metabolites across both time-points associated with exposure to any overnutrition, using a false-discovery-rate correction (FDR) <0.20. These metabolites were included in a principal component analysis (PCA) to generate profiles and assess metabolite profile differences with respect to overnutrition typology (adjusted for prenatal smoking, offspring age, sex, and race/ethnicity). Fetal overnutrition was associated with 52 metabolites. PCA yielded four factors accounting for 17−27% of the variance, depending on age of measurement. We observed differences in three factor patterns with respect to overnutrition typology: sphingomyelin-mannose (8−13% variance), skeletal muscle metabolism (6−10% variance), and 3-carboxy-4-methyl-5-propyl-2-furanpropanoic acid (CMPF; 3−4% variance). The sphingomyelin-mannose factor score was higher among offspring exposed to obesity vs. GDM. Exposure to obesity + GDM (vs. GDM or obesity only) was associated with higher skeletal muscle metabolism and CMPF scores. Fetal overnutrition is associated with metabolic changes in the offspring, but differences between typologies of overnutrition account for a small amount of variation in the metabolome, suggesting there is likely greater pathophysiological overlap than difference.
Collapse
Affiliation(s)
- Ellen C. Francis
- Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, University of Colorado Denver Anschutz Medical Campus, Aurora, CO 80045, USA; (C.C.C.); (D.D.); (W.P.)
| | - Katerina Kechris
- Department of Biostatistics and Informatics, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA;
| | - Catherine C. Cohen
- Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, University of Colorado Denver Anschutz Medical Campus, Aurora, CO 80045, USA; (C.C.C.); (D.D.); (W.P.)
- Department of Pediatrics, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | | | - Dana Dabelea
- Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, University of Colorado Denver Anschutz Medical Campus, Aurora, CO 80045, USA; (C.C.C.); (D.D.); (W.P.)
- Department of Pediatrics, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Denver Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Wei Perng
- Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, University of Colorado Denver Anschutz Medical Campus, Aurora, CO 80045, USA; (C.C.C.); (D.D.); (W.P.)
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Denver Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
102
|
Zheng Y, Xu Q, Jin Q, Du Y, Yan J, Gao H, Zheng H. Urinary and faecal metabolic characteristics in APP/PS1 transgenic mouse model of Alzheimer's disease with and without cognitive decline. Biochem Biophys Res Commun 2022; 604:130-136. [PMID: 35303679 DOI: 10.1016/j.bbrc.2022.03.048] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 03/09/2022] [Indexed: 11/02/2022]
Abstract
Alzheimer's disease (AD) has been considered to be a systematic metabolic disorder, but little information is available about metabolic changes in the urine and feces. In this study, we investigated urinary and faecal metabolic profiles in amyloid precursor protein/presenilin 1 (APP/PS1) mice at 3 and 9 months of age (3 M and 9 M) and age-matched wild-type (WT) mice by using 1H NMR-based metabolomics, and aimed to explore changes in metabolic pathways during amyloid pathology progression and identify potential metabolite biomarkers at earlier stage of AD. The results show that learning and memory abilities were impaired in APP/PS1 mice relative to WT mice at 9 M, but not at 3 M. However, metabolomics analysis demonstrates that AD disrupted metabolic phenotypes in the urine and feces of APP/PS1 mice at both 3 M and 9 M, including amino acid metabolism, microbial metabolism and energy metabolism. In addition, several potential metabolite biomarkers were identified for discriminating AD and WT mice prior to cognitive decline with the AUC values from 0.755 to 0.971, such as taurine, hippurate, urea and methylamine in the urine as well as alanine, leucine and valine in the feces. Therefore, our results not only confirmed AD as a metabolic disorder, but also contributed to the identification of potential biomarkers at earlier stage of AD.
Collapse
Affiliation(s)
- Yafei Zheng
- Institute of Metabonomics & Medical NMR, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Qingqing Xu
- Institute of Metabonomics & Medical NMR, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Qihao Jin
- Institute of Metabonomics & Medical NMR, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Yao Du
- Institute of Metabonomics & Medical NMR, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Junjie Yan
- Institute of Metabonomics & Medical NMR, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Hongchang Gao
- Institute of Metabonomics & Medical NMR, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China.
| | - Hong Zheng
- Institute of Metabonomics & Medical NMR, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China.
| |
Collapse
|
103
|
Martin WP, Malmodin D, Pedersen A, Wallace M, Fändriks L, Aboud CM, Petry TBZ, Cunha da Silveira LP, da Costa Silva ACC, Cohen RV, le Roux CW, Docherty NG. Urinary Metabolomic Changes Accompanying Albuminuria Remission following Gastric Bypass Surgery for Type 2 Diabetic Kidney Disease. Metabolites 2022; 12:139. [PMID: 35186675 PMCID: PMC7612403 DOI: 10.3390/metabo12020139] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
In the Microvascular Outcomes after Metabolic Surgery randomised clinical trial (MOMS RCT, NCT01821508), combined metabolic surgery (gastric bypass) plus medical therapy (CSM) was superior to medical therapy alone (MTA) as a means of achieving albuminuria remission at 2-year follow-up in patients with obesity and early diabetic kidney disease (DKD). In the present study, we assessed the urinary 1H-NMR metabolome in a subgroup of patients from both arms of the MOMS RCT at baseline and 6-month follow-up. Whilst CSM and MTA both reduced the urinary excretion of sugars, CSM generated a distinctive urinary metabolomic profile characterised by increases in host–microbial co-metabolites (N-phenylacetylglycine, trimethylamine N-oxide, and 4-aminobutyrate (GABA)) and amino acids (arginine and glutamine). Furthermore, reductions in aromatic amino acids (phenylalanine and tyrosine), as well as branched-chain amino acids (BCAAs) and related catabolites (valine, leucine, 3-hydroxyisobutyrate, 3-hydroxyisovalerate, and 3-methyl-2-oxovalerate), were observed following CSM but not MTA. Improvements in BMI did not correlate with improvements in metabolic and renal indices following CSM. Conversely, urinary metabolites changed by CSM at 6 months were moderately to strongly correlated with improvements in blood pressure, glycaemia, triglycerides, and albuminuria up to 24 months following treatment initiation, highlighting the potential involvement of these shifts in the urinary metabolomic profile in the metabolic and renoprotective effects of CSM.
Collapse
Affiliation(s)
- William P. Martin
- Diabetes Complications Research Centre, School of Medicine, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, D04 V1W8 Dublin, Ireland; (W.P.M.); (C.W.l.R.)
| | - Daniel Malmodin
- Swedish NMR Centre, University of Gothenburg, 40530 Gothenburg, Sweden; (D.M.); (A.P.)
| | - Anders Pedersen
- Swedish NMR Centre, University of Gothenburg, 40530 Gothenburg, Sweden; (D.M.); (A.P.)
| | - Martina Wallace
- Institute of Food and Health, School of Agriculture and Food Science, University College Dublin, Belfield, D04 V1W8 Dublin, Ireland;
| | - Lars Fändriks
- Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, 40530 Gothenburg, Sweden;
| | - Cristina M. Aboud
- The Centre for Obesity and Diabetes, Oswaldo Cruz German Hospital, São Paulo 01333-010, Brazil; (C.M.A.); (T.B.Z.P.); (L.P.C.d.S.); (A.C.C.d.C.S.); (R.V.C.)
| | - Tarissa B. Zanata Petry
- The Centre for Obesity and Diabetes, Oswaldo Cruz German Hospital, São Paulo 01333-010, Brazil; (C.M.A.); (T.B.Z.P.); (L.P.C.d.S.); (A.C.C.d.C.S.); (R.V.C.)
| | - Lívia P. Cunha da Silveira
- The Centre for Obesity and Diabetes, Oswaldo Cruz German Hospital, São Paulo 01333-010, Brazil; (C.M.A.); (T.B.Z.P.); (L.P.C.d.S.); (A.C.C.d.C.S.); (R.V.C.)
| | - Ana C. Calmon da Costa Silva
- The Centre for Obesity and Diabetes, Oswaldo Cruz German Hospital, São Paulo 01333-010, Brazil; (C.M.A.); (T.B.Z.P.); (L.P.C.d.S.); (A.C.C.d.C.S.); (R.V.C.)
| | - Ricardo V. Cohen
- The Centre for Obesity and Diabetes, Oswaldo Cruz German Hospital, São Paulo 01333-010, Brazil; (C.M.A.); (T.B.Z.P.); (L.P.C.d.S.); (A.C.C.d.C.S.); (R.V.C.)
| | - Carel W. le Roux
- Diabetes Complications Research Centre, School of Medicine, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, D04 V1W8 Dublin, Ireland; (W.P.M.); (C.W.l.R.)
- Diabetes Research Group, Ulster University, Coleraine BT52 1SA, UK
| | - Neil G. Docherty
- Diabetes Complications Research Centre, School of Medicine, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, D04 V1W8 Dublin, Ireland; (W.P.M.); (C.W.l.R.)
- Correspondence:
| |
Collapse
|
104
|
He Y, Zhang H, Yang Y, Yu X, Zhang X, Xing Q, Zhang G. Using Metabolomics in Diabetes Management with Traditional Chinese Medicine: A Review. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2022; 49:1813-1837. [PMID: 34961417 DOI: 10.1142/s0192415x21500865] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The incidence of diabetes worldwide continues to rise, placing a huge economic and medical burden on human society. More than 90% of diabetic cases are type 2 diabetes (T2D). At present, the pathogenesis of T2D is not yet fully understood. Metabolomics uses high-resolution analytical techniques (typically NMR and MS) to help identify biomarkers associated with the risk of T2D and reveal potential pathogenesis. Many metabolites such as branched-chain amino acids (BCAAs), aromatic amino acids, glycine, 2-hydroxybutyric acid (2-HB), lysophosphatidylcholine (LPC) (18:2), and trehalose have proven to be biomarkers of T2D. Insulin resistance (IR) induced by BCAA in T2D mice is related to the activation of mammalian target of rapamycin (mTOR) and phosphorylation of insulin receptor substrate-1 (IRS1). Incomplete LCFA [Formula: see text]-oxidation promote acylcarnitine byproduct accumulation and stimulates proinflammatory NF[Formula: see text]B-related pathways to inhibit insulin action. Traditional Chinese Medicine (TCM) presents unique advantages in the treatment of T2D. Multiple metabolites and metabolic pathways have been identified in the treatment of TCM, providing valuable biomarkers and novel targets for drug therapy and pharmacological mechanism. Therefore, this paper reviews the modern achievements of metabolomics in T2D research and the progress of TCM management in recent years, in order to provide valuable information for related research.
Collapse
Affiliation(s)
- Yanling He
- Graduate School of Hebei University of Traditional, Chinese Medicine, Shijiazhuang 050091, P. R. China
| | - Hefang Zhang
- Graduate School of Hebei University of Traditional, Chinese Medicine, Shijiazhuang 050091, P. R. China.,Department of Endocrinology, First Affiliated Hospital of Hebei University of Traditional, Chinese Medicine, Shijiazhuang 050011, P. R. China
| | - Yufei Yang
- Graduate School of Hebei University of Traditional, Chinese Medicine, Shijiazhuang 050091, P. R. China
| | - Xianghui Yu
- Department of Endocrinology, First Affiliated Hospital of Hebei University of Traditional, Chinese Medicine, Shijiazhuang 050011, P. R. China
| | - Xiao Zhang
- Graduate School of Hebei University of Traditional, Chinese Medicine, Shijiazhuang 050091, P. R. China
| | - Qiaolin Xing
- Graduate School of Hebei University of Traditional, Chinese Medicine, Shijiazhuang 050091, P. R. China
| | - Gengliang Zhang
- Graduate School of Hebei University of Traditional, Chinese Medicine, Shijiazhuang 050091, P. R. China.,Department of Endocrinology, First Affiliated Hospital of Hebei University of Traditional, Chinese Medicine, Shijiazhuang 050011, P. R. China
| |
Collapse
|
105
|
He Q, Dong H, Guo Y, Gong M, Xia Q, Lu F, Wang D. Multi-target regulation of intestinal microbiota by berberine to improve type 2 diabetes mellitus. Front Endocrinol (Lausanne) 2022; 13:1074348. [PMID: 36465656 PMCID: PMC9715767 DOI: 10.3389/fendo.2022.1074348] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 11/07/2022] [Indexed: 11/21/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) and its complications are major public health problems that seriously affect the quality of human life. The modification of intestinal microbiota has been widely recognized for the management of diabetes. The relationship between T2DM, intestinal microbiota, and active ingredient berberine (BBR) in intestinal microbiota was reviewed in this paper. First of all, the richness and functional changes of intestinal microbiota disrupt the intestinal environment through the destruction of the intestinal barrier and fermentation/degradation of pathogenic/protective metabolites, targeting the liver, pancreas, visceral adipose tissue (VAT), etc., to affect intestinal health, blood glucose, and lipids, insulin resistance and inflammation. Then, we focus on BBR, which protects the composition of intestinal microbiota, the changes of intestinal metabolites, and immune regulation disorder of the intestinal environment as the therapeutic mechanism as well as its current clinical trials. Further research can analyze the mechanism network of BBR to exert its therapeutic effect according to its multi-target compound action, to provide a theoretical basis for the use of different phytochemical components alone or in combination to prevent and treat T2DM or other metabolic diseases by regulating intestinal microbiota.
Collapse
Affiliation(s)
- Qiongyao He
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hui Dong
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yujin Guo
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Minmin Gong
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Qingsong Xia
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Fuer Lu
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
- *Correspondence: Fuer Lu, ; Dingkun Wang,
| | - Dingkun Wang
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
- *Correspondence: Fuer Lu, ; Dingkun Wang,
| |
Collapse
|
106
|
Liu L, Zhang J, Cheng Y, Zhu M, Xiao Z, Ruan G, Wei Y. Gut microbiota: A new target for T2DM prevention and treatment. Front Endocrinol (Lausanne) 2022; 13:958218. [PMID: 36034447 PMCID: PMC9402911 DOI: 10.3389/fendo.2022.958218] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 07/22/2022] [Indexed: 12/12/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM), one of the fastest growing metabolic diseases, has been characterized by metabolic disorders including hyperglycemia, hyperlipidemia and insulin resistance (IR). In recent years, T2DM has become the fastest growing metabolic disease in the world. Studies have indicated that patients with T2DM are often associated with intestinal flora disorders and dysfunction involving multiple organs. Metabolites of the intestinal flora, such as bile acids (BAs), short-chain fatty acids (SCFAs) and amino acids (AAs)may influence to some extent the decreased insulin sensitivity associated with T2DM dysfunction and regulate metabolic as well as immune homeostasis. In this paper, we review the changes in the gut flora in T2DM and the mechanisms by which the gut microbiota modulates metabolites affecting T2DM, which may provide a basis for the early identification of T2DM-susceptible individuals and guide targeted interventions. Finally, we also highlight gut microecological therapeutic strategies focused on shaping the gut flora to inform the improvement of T2DM progression.
Collapse
Affiliation(s)
- Lulu Liu
- Department of Gastroenterology, Chongqing Key Laboratory of Digestive Malignancies, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
- Department of Plastic and Cosmetic Surgery, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Jiheng Zhang
- Department of Plastic and Cosmetic Surgery, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Yi Cheng
- Department of Gastroenterology, Chongqing Key Laboratory of Digestive Malignancies, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Meng Zhu
- Department of Plastic and Cosmetic Surgery, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Zhifeng Xiao
- Department of Gastroenterology, Chongqing Key Laboratory of Digestive Malignancies, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Guangcong Ruan
- Department of Gastroenterology, Chongqing Key Laboratory of Digestive Malignancies, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
- *Correspondence: Yanling Wei, ; Guangcong Ruan,
| | - Yanling Wei
- Department of Gastroenterology, Chongqing Key Laboratory of Digestive Malignancies, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
- *Correspondence: Yanling Wei, ; Guangcong Ruan,
| |
Collapse
|
107
|
Yavuz E, Irak K, Çelik ÖY, Bolacali M, Ergiden Y, Gürgöze S. Determination of the Relationship of Serum Amino Acid Profile with Sex and Body Weight in Healthy Geese by Liquid Chromatography-Tandem Mass Spectrometry. BRAZILIAN JOURNAL OF POULTRY SCIENCE 2022. [DOI: 10.1590/1806-9061-2021-1569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- E Yavuz
- International Center for Livestock Research and Training, Turkey
| | | | | | | | - Y Ergiden
- International Center for Livestock Research and Training, Turkey
| | | |
Collapse
|
108
|
Zhou C, Zhang Q, Lu L, Wang J, Liu D, Liu Z. Metabolomic Profiling of Amino Acids in Human Plasma Distinguishes Diabetic Kidney Disease From Type 2 Diabetes Mellitus. Front Med (Lausanne) 2021; 8:765873. [PMID: 34912824 PMCID: PMC8666657 DOI: 10.3389/fmed.2021.765873] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 11/01/2021] [Indexed: 01/02/2023] Open
Abstract
Background: Diabetic kidney disease (DKD) is a highly prevalent complication in patients with type 2 diabetes mellitus (T2DM). Patients with DKD exhibit changes in plasma levels of amino acids (AAs) due to insulin resistance, reduced protein intake, and impaired renal transport of AAs. The role of AAs in distinguishing DKD from T2DM and healthy controls has yet to be elucidated. This study aimed to investigate the metabolomic profiling of AAs in the plasma of patients with DKD. Methods: We established an ultra-performance liquid chromatography tandem mass spectrometry (UPLC-MS/MS) method to detect the plasma levels of the 20 AAs in healthy controls (n = 112), patients with T2DM (n = 101), and patients with DKD (n = 101). The key AAs associated with DKD were identified by orthogonal partial least-squares discriminant analysis (OPLS-DA) models with loading plots, shared and unique structures (SUS) plots, and variable importance in projection (VIP) values. The discrimination accuracies of these key AAs were then determined by analyses of receiver-operating characteristic (ROC) curves. Results: Metabolomic profiling of plasma revealed significant alterations in levels of the 20 AAs in patients with DKD when compared to those in either patients with T2DM or healthy controls. Metabolomic profiling of the 20 AAs showed a visual separation of patients with DKD from patients with T2DM and healthy controls in OPLS-DA models. Based on loading plots, SUS plots, and VIP values in the OPLS-DA models, we identified valine and cysteine as potential contributors to the progression of DKD from patients with T2DM. Histidine was identified as a key mediator that could distinguish patients with DKD from healthy controls. Plasma levels of histidine and valine were decreased significantly in patients with DKD with a decline in kidney function, and had excellent performance in distinguishing patients with DKD from patients with T2DM and healthy controls according to ROC curves. Conclusion: Plasma levels of histidine and valine were identified as the main AAs that can distinguish patients with DKD. Our findings provide new options for the prevention, treatment, and management of DKD.
Collapse
Affiliation(s)
- Chunyu Zhou
- Blood Purification Center, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Research Institute of Nephrology, Zhengzhou University, Zhengzhou, China
| | - Qing Zhang
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, China.,Department of Nephrology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Liqian Lu
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, China.,Department of Nephrology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jiao Wang
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, China.,Department of Nephrology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Dongwei Liu
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, China.,Department of Nephrology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Zhangsuo Liu
- Blood Purification Center, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Research Institute of Nephrology, Zhengzhou University, Zhengzhou, China.,Department of Nephrology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| |
Collapse
|
109
|
Gander J, Carrard J, Gallart-Ayala H, Borreggine R, Teav T, Infanger D, Colledge F, Streese L, Wagner J, Klenk C, Nève G, Knaier R, Hanssen H, Schmidt-Trucksäss A, Ivanisevic J. Metabolic Impairment in Coronary Artery Disease: Elevated Serum Acylcarnitines Under the Spotlights. Front Cardiovasc Med 2021; 8:792350. [PMID: 34977199 PMCID: PMC8716394 DOI: 10.3389/fcvm.2021.792350] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 11/09/2021] [Indexed: 12/26/2022] Open
Abstract
Coronary artery disease (CAD) remains the leading cause of death worldwide. Expanding patients' metabolic phenotyping beyond clinical chemistry investigations could lead to earlier recognition of disease onset and better prevention strategies. Additionally, metabolic phenotyping, at the molecular species level, contributes to unravel the roles of metabolites in disease development. In this cross-sectional study, we investigated clinically healthy individuals (n = 116, 65% male, 70.8 ± 8.7 years) and patients with CAD (n = 54, 91% male, 67.0 ± 11.5 years) of the COmPLETE study. We applied a high-coverage quantitative liquid chromatography-mass spectrometry approach to acquire a comprehensive profile of serum acylcarnitines, free carnitine and branched-chain amino acids (BCAAs), as markers of mitochondrial health and energy homeostasis. Multivariable linear regression analyses, adjusted for confounders, were conducted to assess associations between metabolites and CAD phenotype. In total, 20 short-, medium- and long-chain acylcarnitine species, along with L-carnitine, valine and isoleucine were found to be significantly (adjusted p ≤ 0.05) and positively associated with CAD. For 17 acylcarnitine species, associations became stronger as the number of affected coronary arteries increased. This implies that circulating acylcarnitine levels reflect CAD severity and might play a role in future patients' stratification strategies. Altogether, CAD is characterized by elevated serum acylcarnitine and BCAA levels, which indicates mitochondrial imbalance between fatty acid and glucose oxidation.
Collapse
Affiliation(s)
- Joséphine Gander
- Division of Sports and Exercise Medicine, Department of Sport, Exercise and Health, University of Basel, Basel, Switzerland
| | - Justin Carrard
- Division of Sports and Exercise Medicine, Department of Sport, Exercise and Health, University of Basel, Basel, Switzerland
| | - Hector Gallart-Ayala
- Metabolomics Platform, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Rébecca Borreggine
- Metabolomics Platform, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Tony Teav
- Metabolomics Platform, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Denis Infanger
- Division of Sports and Exercise Medicine, Department of Sport, Exercise and Health, University of Basel, Basel, Switzerland
| | - Flora Colledge
- Division of Sports Science, Department of Sport, Exercise and Health, University of Basel, Basel, Switzerland
| | - Lukas Streese
- Division of Sports and Exercise Medicine, Department of Sport, Exercise and Health, University of Basel, Basel, Switzerland
| | - Jonathan Wagner
- Division of Sports and Exercise Medicine, Department of Sport, Exercise and Health, University of Basel, Basel, Switzerland
| | - Christopher Klenk
- Division of Sports and Exercise Medicine, Department of Sport, Exercise and Health, University of Basel, Basel, Switzerland
| | - Gilles Nève
- Division of Sports and Exercise Medicine, Department of Sport, Exercise and Health, University of Basel, Basel, Switzerland
| | - Raphael Knaier
- Division of Sports and Exercise Medicine, Department of Sport, Exercise and Health, University of Basel, Basel, Switzerland
| | - Henner Hanssen
- Division of Sports and Exercise Medicine, Department of Sport, Exercise and Health, University of Basel, Basel, Switzerland
| | - Arno Schmidt-Trucksäss
- Division of Sports and Exercise Medicine, Department of Sport, Exercise and Health, University of Basel, Basel, Switzerland
- Arno Schmidt-Trucksäss
| | - Julijana Ivanisevic
- Metabolomics Platform, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
- *Correspondence: Julijana Ivanisevic
| |
Collapse
|
110
|
Chevli PA, Freedman BI, Hsu FC, Xu J, Rudock ME, Ma L, Parks JS, Palmer ND, Shapiro MD. Plasma metabolomic profiling in subclinical atherosclerosis: the Diabetes Heart Study. Cardiovasc Diabetol 2021; 20:231. [PMID: 34876126 PMCID: PMC8653597 DOI: 10.1186/s12933-021-01419-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 11/15/2021] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND Incidence rates of cardiovascular disease (CVD) are increasing, partly driven by the diabetes epidemic. Novel prediction tools and modifiable treatment targets are needed to enhance risk assessment and management. Plasma metabolite associations with subclinical atherosclerosis were investigated in the Diabetes Heart Study (DHS), a cohort enriched for type 2 diabetes (T2D). METHODS The analysis included 700 DHS participants, 438 African Americans (AAs), and 262 European Americans (EAs), in whom coronary artery calcium (CAC) was assessed using ECG-gated computed tomography. Plasma metabolomics using liquid chromatography-mass spectrometry identified 853 known metabolites. An ancestry-specific marginal model incorporating generalized estimating equations examined associations between metabolites and CAC (log-transformed (CAC + 1) as outcome measure). Models were adjusted for age, sex, BMI, diabetes duration, date of plasma collection, time between plasma collection and CT exam, low-density lipoprotein cholesterol (LDL-C), and statin use. RESULTS At an FDR-corrected p-value < 0.05, 33 metabolites were associated with CAC in AAs and 36 in EAs. The androgenic steroids, fatty acid, phosphatidylcholine, and bile acid metabolism subpathways were associated with CAC in AAs, whereas fatty acid, lysoplasmalogen, and branched-chain amino acid (BCAA) subpathways were associated with CAC in EAs. CONCLUSIONS Strikingly different metabolic signatures were associated with subclinical coronary atherosclerosis in AA and EA DHS participants.
Collapse
Affiliation(s)
- Parag Anilkumar Chevli
- Section on Hospital Medicine, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Barry I Freedman
- Section on Nephrology, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Fang-Chi Hsu
- Department of Biostatistics and Data Science, Division of Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Jianzhao Xu
- Department of Biochemistry, Wake Forest School of Medicine, 1 Medical Center Blvd, Winston-Salem, NC, 27157, USA
| | - Megan E Rudock
- Department of Biochemistry, Wake Forest School of Medicine, 1 Medical Center Blvd, Winston-Salem, NC, 27157, USA
| | - Lijun Ma
- Section on Nephrology, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - John S Parks
- Section on Molecular Medicine, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Nicholette D Palmer
- Department of Biochemistry, Wake Forest School of Medicine, 1 Medical Center Blvd, Winston-Salem, NC, 27157, USA.
| | - Michael D Shapiro
- Section of Cardiovascular Medicine, Center for Preventive Cardiology, Wake Forest School of Medicine, 1 Medical Center Blvd, Winston-Salem, NC, 27157, USA.
| |
Collapse
|
111
|
Taya N, Katakami N, Omori K, Arakawa S, Hosoe S, Watanabe H, Takahara M, Miyashita K, Nishizawa H, Matsuoka T, Furuno M, Bamba T, Iida J, Fukusaki E, Shimomura I. Evaluation of change in metabolome caused by comprehensive diabetes treatment: A prospective observational study of diabetes inpatients with gas chromatography/mass spectrometry-based non-target metabolomic analysis. J Diabetes Investig 2021; 12:2232-2241. [PMID: 34032389 PMCID: PMC8668060 DOI: 10.1111/jdi.13600] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 04/17/2021] [Accepted: 05/20/2021] [Indexed: 11/28/2022] Open
Abstract
AIMS/INTRODUCTION Diabetes patients develop a variety of metabolic abnormalities in addition to hyperglycemia. However, details regarding change in various metabolites after comprehensive diabetes treatment remain unknown. This study aimed to identify the short-term change in metabolome in inpatients who were subject to comprehensive diabetes treatment, using gas chromatography/mass spectrometry-based non-target metabolomics techniques. MATERIALS AND METHODS Participants of the present study were randomly recruited from the patients with type 2 diabetes hospitalized due to problems with glycemic control (n = 31) and volunteers without diabetes (n = 30), both of whom were aged between 20 and 75 years. A metabolomic analysis of fasting plasma samples on the 2nd (pre-treatment) and 16th hospital (post-treatment) day with gas chromatography/mass spectrometry using a multiple reaction monitoring mode was carried out. RESULTS A principal component analysis showed that metabolome of fasting plasma was different between individuals with and without diabetes. The metabolome of fasting plasma in diabetes patients after treatment was different from that of pre-treatment, as well as individuals without diabetes. Many amino acids (proline, glycine, serine, threonine, methionine, pyroglutamic acid, glutamine and lysine) were significantly increased by >10% after administering the inpatient diabetes treatment. A hierarchical clustering analysis showed that in the case of patients with markedly decreased monosaccharide levels and increased 1,5-anhydroglucitol, the levels of amino acids increased more significantly. CONCLUSIONS After a 2-week comprehensive treatment, the plasma levels of various amino acids increased in conjunction with the reduction in monosaccharide levels in poorly controlled type 2 diabetes patients.
Collapse
Affiliation(s)
- Naohiro Taya
- Department of Metabolic MedicineOsaka University Graduate School of MedicineOsakaJapan
| | - Naoto Katakami
- Department of Metabolic MedicineOsaka University Graduate School of MedicineOsakaJapan
- Department of Metabolism and AtherosclerosisOsaka University Graduate School of MedicineOsakaJapan
| | - Kazuo Omori
- Department of Metabolic MedicineOsaka University Graduate School of MedicineOsakaJapan
| | - Shoya Arakawa
- Laboratory of Bioresource EngineeringDepartment of BiotechnologyGraduate School of EngineeringOsaka UniversityOsakaJapan
| | - Shigero Hosoe
- Department of Metabolic MedicineOsaka University Graduate School of MedicineOsakaJapan
| | - Hirotaka Watanabe
- Department of Metabolic MedicineOsaka University Graduate School of MedicineOsakaJapan
| | - Mitsuyoshi Takahara
- Department of Metabolic MedicineOsaka University Graduate School of MedicineOsakaJapan
- Department of Diabetes Care MedicineGraduate School of MedicineOsaka UniversityOsakaJapan
| | - Kazuyuki Miyashita
- Department of Metabolic MedicineOsaka University Graduate School of MedicineOsakaJapan
| | - Hitoshi Nishizawa
- Department of Metabolic MedicineOsaka University Graduate School of MedicineOsakaJapan
| | - Taka‐Aki Matsuoka
- Department of Metabolic MedicineOsaka University Graduate School of MedicineOsakaJapan
| | - Masahiro Furuno
- Laboratory of Bioresource EngineeringDepartment of BiotechnologyGraduate School of EngineeringOsaka UniversityOsakaJapan
| | - Takeshi Bamba
- Division of MetabolomicsResearch Center for Transomics MedicineMedical Institute of BioregulationKyushu UniversityFukuokaJapan
| | - Junko Iida
- Shimadzu CorporationKyotoJapan
- Osaka University Shimadzu Omics Innovation Research LaboratoriesGraduate School of EngineeringOsaka UniversityOsakaJapan
| | - Eiichiro Fukusaki
- Laboratory of Bioresource EngineeringDepartment of BiotechnologyGraduate School of EngineeringOsaka UniversityOsakaJapan
| | - Iichiro Shimomura
- Department of Metabolic MedicineOsaka University Graduate School of MedicineOsakaJapan
| |
Collapse
|
112
|
Mastrototaro L, Roden M. Insulin resistance and insulin sensitizing agents. Metabolism 2021; 125:154892. [PMID: 34563556 DOI: 10.1016/j.metabol.2021.154892] [Citation(s) in RCA: 87] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 09/08/2021] [Accepted: 09/20/2021] [Indexed: 02/06/2023]
Abstract
Insulin resistance is a common feature of obesity and type 2 diabetes, but novel approaches of diabetes subtyping (clustering) revealed variable degrees of insulin resistance in people with diabetes. Specifically, the severe insulin resistant diabetes (SIRD) subtype not only exhibits metabolic abnormalities, but also bears a higher risk for cardiovascular, renal and hepatic comorbidities. In humans, insulin resistance comprises dysfunctional adipose tissue, lipotoxic insulin signaling followed by glucotoxicity, oxidative stress and low-grade inflammation. Recent studies show that aside from metabolites (free fatty acids, amino acids) and signaling proteins (myokines, adipokines, hepatokines) also exosomes with their cargo (proteins, mRNA and microRNA) contribute to altered crosstalk between skeletal muscle, liver and adipose tissue during the development of insulin resistance. Reduction of fat mass mainly, but not exclusively, explains the success of lifestyle modification and bariatric surgery to improve insulin sensitivity. Moreover, some older antihyperglycemic drugs (metformin, thiazolidinediones), but also novel therapeutic concepts (new peroxisome proliferator-activated receptor agonists, incretin mimetics, sodium glucose cotransporter inhibitors, modulators of energy metabolism) can directly or indirectly reduce insulin resistance. This review summarizes molecular mechanisms underlying insulin resistance including the roles of exosomes and microRNAs, as well as strategies for the management of insulin resistance in humans.
Collapse
Affiliation(s)
- Lucia Mastrototaro
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine University Düsseldorf, Düsseldorf, Germany; German Center for Diabetes Research, Partner Düsseldorf, München-Neuherberg, Germany
| | - Michael Roden
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine University Düsseldorf, Düsseldorf, Germany; German Center for Diabetes Research, Partner Düsseldorf, München-Neuherberg, Germany; Department of Endocrinology and Diabetology, Medical Faculty and University Hospital, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany.
| |
Collapse
|
113
|
Zhang L, Zhang Y, Ma Z, Zhu Y, Chen Z. Altered amino acid metabolism between coronary heart disease patients with and without type 2 diabetes by quantitative 1H NMR based metabolomics. J Pharm Biomed Anal 2021; 206:114381. [PMID: 34583124 DOI: 10.1016/j.jpba.2021.114381] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 08/30/2021] [Accepted: 09/15/2021] [Indexed: 01/01/2023]
Abstract
Coronary heart disease (CHD) is a multifactorial disease associated with complicated altered metabolic pathways, especially when it occurs with type 2 diabetes (T2D). Identification of metabolic changes could provide further understanding of the underlying pathological link between with CHD and T2D. In this study, 61 controls, 30 consecutive patients with only T2D, 188 with confirmed CHD among which 50 also having T2D were enrolled. Quantitative 1H nuclear magnetic resonance (1H NMR) based metabonomic approach was used to obtain the plasma metabolic profiles of the study samples. Multivariate principal component analysis and orthogonal partial least-squares discriminant analysis (OPLS-DA) were used to evaluate the differences of metabolites. Compared with controls, there were significant differences in CHD patient with T2D groups including BCAAs (isoleucine, leucine, and valine) and phenylalanine by using quantitative metabolomic profiling (all p < 0.05). With OPLS-DA, the results showed that, compared to CHD patients without T2D, there had higher levels of metabolites for leucine, valine, fumarate, tyrosine, and phenylalanine in the plasma of CHD patients with T2D. Moreover, the CHD patients with T2D showed modified phenylalanine, tyrosine and tryptophan biosynthesis, valine, leucine and isoleucine biosynthesis and degradation compared with CHD patients without T2D. By 1H NMR based quantitative metabolomic method, our findings suggest that T2D has important effects on BCAAs and AAAs metabolism in CHD patients indicating altered cardiac energy metabolism and activated signaling pathway could be potential targets for therapy in CHD disease combined with T2D.
Collapse
Affiliation(s)
- Lili Zhang
- Department of Cardiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China.
| | - Yifan Zhang
- Department of Cardiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China.
| | - Zhihui Ma
- Department of Cardiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China.
| | - Yanrong Zhu
- Department of Cardiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China.
| | - Zhong Chen
- Department of Cardiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China.
| |
Collapse
|
114
|
Zhang H, Zhao Y, Zhao D, Chen X, Khan NU, Liu X, Zheng Q, Liang Y, Zhu Y, Iqbal J, Lin J, Shen L. Potential biomarkers identified in plasma of patients with gestational diabetes mellitus. Metabolomics 2021; 17:99. [PMID: 34739593 DOI: 10.1007/s11306-021-01851-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 10/29/2021] [Indexed: 12/26/2022]
Abstract
Gestational diabetes mellitus (GDM) is a common complication during pregnancy. Looking for reliable diagnostic markers for early diagnosis can reduce the impact of the disease on the fetus OBJECTIVE: The present study is designed to find plasma metabolites that can be used as potential biomarkers for GDM, and to clarify GDM-related mechanisms METHODS: By non-target metabolomics analysis, compared with their respective controls, the plasma metabolites of GDM pregnant women at 12-16 weeks and 24-28 weeks of pregnancy were analyzed. Multiple reaction monitoring (MRM) analysis was performed to verify the potential marker RESULTS: One hundred and seventy-two (172) and 478 metabolites were identified as differential metabolites in the plasma of GDM pregnant women at 12-16 weeks and 24-28 weeks of pregnancy, respectively. Among these, 40 metabolites were overlapped. Most of them are associated with the mechanism of diabetes, and related to short-term and long-term complications in the perinatal period. Among them, 7 and 10 differential metabolites may serve as potential biomarkers at the 12-16 weeks and 24-28 weeks of pregnancy, respectively. By MRM analysis, compared with controls, increased levels of 17(S)-HDoHE and sebacic acid may serve as early prediction biomarkers of GDM. At 24-28 weeks of pregnancy, elevated levels of 17(S)-HDoHE and L-Serine may be used as auxiliary diagnostic markers for GDM CONCLUSION: Abnormal amino acid metabolism and lipid metabolism in patients with GDM may be related to GDM pathogenesis. Several differential metabolites identified in this study may serve as potential biomarkers for GDM prediction and diagnosis.
Collapse
Affiliation(s)
- Huajie Zhang
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, 518071, People's Republic of China
| | - Yuxi Zhao
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, 518071, People's Republic of China
| | - Danqing Zhao
- Department of Obstetrics and Gynecology, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, People's Republic of China
| | - Xinqian Chen
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, 518071, People's Republic of China
| | - Naseer Ullah Khan
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, 518071, People's Republic of China
| | - Xukun Liu
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, 518071, People's Republic of China
| | - Qihong Zheng
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, 518071, People's Republic of China
| | - Yi Liang
- Department of Obstetrics and Gynecology, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, People's Republic of China
| | - Yuhua Zhu
- Department of Obstetrics and Gynecology, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, People's Republic of China
| | - Javed Iqbal
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, 518071, People's Republic of China
| | - Jing Lin
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, 518071, People's Republic of China
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, Shenzhen, 518071, People's Republic of China
| | - Liming Shen
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, 518071, People's Republic of China.
- Brain Disease and Big Data Research Institute, Shenzhen University, Shenzhen, 518071, People's Republic of China.
| |
Collapse
|
115
|
Neacsu M, Vaughan NJ, Multari S, Haljas E, Scobbie L, Duncan GJ, Cantlay L, Fyfe C, Anderson S, Horgan G, Johnstone AM, Russell WR. Hemp and buckwheat are valuable sources of dietary amino acids, beneficially modulating gastrointestinal hormones and promoting satiety in healthy volunteers. Eur J Nutr 2021; 61:1057-1072. [PMID: 34716790 PMCID: PMC8854285 DOI: 10.1007/s00394-021-02711-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 10/13/2021] [Indexed: 12/25/2022]
Abstract
Purpose This study evaluated the postprandial effects following consumption of buckwheat, fava bean, pea, hemp and lupin compared to meat (beef); focussing on biomarkers of satiety, gut hormones, aminoacids and plant metabolites bioavailability and metabolism. Methods Ten subjects (n = 3 men; n = 7 women; 42 ± 11.8 years of age; BMI 26 ± 5.8 kg/m2) participated in six 1-day independent acute interventions, each meal containing 30 g of protein from buckwheat, fava bean, pea, hemp, lupin and meat (beef). Blood samples were collected during 24-h and VAS questionnaires over 5-h. Results Volunteers consumed significantly higher amounts of most amino acids from the meat meal, and with few exceptions, postprandial composition of plasma amino acids was not significantly different after consuming the plant-based meals. Buckwheat meal was the most satious (300 min hunger scores, p < 0.05).Significant increase in GLP-1 plasma (AUC, iAUC p = 0.01) found after hemp compared with the other plant-based meals. Decreased plasma ghrelin concentrations (iAUC p < 0.05) found on plant (hemp) vs. meat meal. Several plasma metabolites after hemp meal consumption were associated with hormone trends (partial least squares-discriminant analysis (PLS-DA): 4-hydroxyphenylpyruvic acid, indole 3-pyruvic acid, 5-hydoxytryptophan, genistein and biochanin A with GLP-1, PYY and insulin; 3-hydroxymandelic acid and luteolidin with GLP-1 and ghrelin and 4-hydroxymandelic acid, benzoic acid and secoisolariciresinol with insulin and ghrelin. Plasma branched-chain amino acids (BCAAs), (iAUC, p < 0.001); and phenylalanine and tyrosine (iAUC, p < 0.05) were lower after buckwheat comparison with meat meal. Conclusion Plants are valuable sources of amino acids which are promoting satiety. The impact of hemp and buckwheat on GLP-1 and, respectively, BCAAs should be explored further as could be relevant for aid and prevention of chronic diseases such as type 2 diabetes. Study registered with clinicaltrial.gov on 12th July 2013, study ID number: NCT01898351. Supplementary Information The online version contains supplementary material available at 10.1007/s00394-021-02711-z.
Collapse
Affiliation(s)
- Madalina Neacsu
- The Rowett Institute, University of Aberdeen, Aberdeen, AB25 2ZD, Scotland, UK.
| | - Nicholas J Vaughan
- The Rowett Institute, University of Aberdeen, Aberdeen, AB25 2ZD, Scotland, UK
| | - Salvatore Multari
- The Rowett Institute, University of Aberdeen, Aberdeen, AB25 2ZD, Scotland, UK
| | - Elisabeth Haljas
- The Rowett Institute, University of Aberdeen, Aberdeen, AB25 2ZD, Scotland, UK
| | - Lorraine Scobbie
- The Rowett Institute, University of Aberdeen, Aberdeen, AB25 2ZD, Scotland, UK
| | - Gary J Duncan
- The Rowett Institute, University of Aberdeen, Aberdeen, AB25 2ZD, Scotland, UK
| | - Louise Cantlay
- The Rowett Institute, University of Aberdeen, Aberdeen, AB25 2ZD, Scotland, UK
| | - Claire Fyfe
- The Rowett Institute, University of Aberdeen, Aberdeen, AB25 2ZD, Scotland, UK
| | - Susan Anderson
- The Rowett Institute, University of Aberdeen, Aberdeen, AB25 2ZD, Scotland, UK
| | - Graham Horgan
- Biomathematics and Statistics Scotland, Aberdeen, AB25 2ZD, Scotland, UK
| | | | - Wendy R Russell
- The Rowett Institute, University of Aberdeen, Aberdeen, AB25 2ZD, Scotland, UK
| |
Collapse
|
116
|
Amino acids predict prognosis in patients with acute dyspnea. BMC Emerg Med 2021; 21:127. [PMID: 34717541 PMCID: PMC8557597 DOI: 10.1186/s12873-021-00519-y] [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: 12/02/2020] [Accepted: 10/19/2021] [Indexed: 11/13/2022] Open
Abstract
Background To identify amino acids that can predict risk of 90-day mortality in patients with acute dyspnea. Method Plasma levels of nine amino acids were analyzed 663 adult patients admitted to the Emergency Department (ED) with acute dyspnea. Cox proportional hazards models were used to examine the relation between amino acid levels and the risk of 90-day mortality. Result Eighty patients (12.1%) died within 90 days of admission. An “Amino Acid Mortality Risk Score” (AMRS), summing absolute plasma levels of glycine, phenylalanine and valine, demonstrated that among the patients belonging to quartile 1 (Q1) of the AMRS, only 4 patients died, compared to 44 patients in quartile 4. Using Q1 of the AMRS as reference, each increment of 1 SD in the AMRS was associated with a hazard ratio (HR) of 2.15 for 90-day mortality, and the HR was > 9 times higher in Q4. Conclusion Glycine, phenylalanine and valine are associated with a risk of 90-day mortality in patients admitted to the ED for acute dyspnea, suggesting that these amino acids may be useful in risk assessments.
Collapse
|
117
|
Circulating amino acids as fingerprints of visceral adipose tissue independent of insulin resistance: a targeted metabolomic research in women. REV ROMANA MED LAB 2021. [DOI: 10.2478/rrlm-2021-0033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Abstract
Introduction: Although obesity and its biomarkers have been intensively studied, little is known about the metabolomic signature of visceral adiposity independent of insulin resistance that frequently accompanies increased levels of visceral fat. Our study aimed to investigate specific changes in amino acid (AA) levels as biomarkers of increased visceral adiposity independent of insulin resistance, in healthy subjects.
Methods: Forty-two adult women were included in this cross-sectional study. Serum samples were analyzed by AAs targeted metabolomics according to their visceral fat area (<100 cm2 and ≥100 cm2).
Results: By corrected t-test and supervised partial least-squares discriminant analysis (PLS-DA) we identified 4 AAs that were significantly higher in the group with higher visceral fat: proline (variable importance in the projection [VIP] predicted value: 1.97), tyrosine (VIP: 2.21), cysteine (VIP: 1.19), isoleucine (VIP: 1.04; p-values <0.05). Also, glycine was significantly lower in the group with higher visceral fat (VIP: 1.65; p-value <0.05). All AAs identified were associated with visceral fat independent of homeo-static model assessment for insulin resistance (p-value for regression coefficients <0.05).
Conclusion: Metabolic pathways that might be disrupted in persons with increased visceral fat are phenylalanine, tyrosine, and tryptophan biosynthesis; tyrosine metabolism; glycine, serine, and threonine metabolism; glyoxylate and dicarboxylate metabolism, and cysteine and methionine metabolism.
Collapse
|
118
|
Abu Bakar Sajak A, Azlan A, Abas F, Hamzah H. The Changes in Endogenous Metabolites in Hyperlipidemic Rats Treated with Herbal Mixture Containing Lemon, Apple Cider, Garlic, Ginger, and Honey. Nutrients 2021; 13:3573. [PMID: 34684574 PMCID: PMC8539352 DOI: 10.3390/nu13103573] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 10/07/2021] [Accepted: 10/09/2021] [Indexed: 02/07/2023] Open
Abstract
An herbal mixture composed of lemon, apple cider, garlic, ginger and honey as a polyphenol-rich mixture (PRM) has been reported to contain hypolipidemic activity on human subjects and hyperlipidemic rats. However, the therapeutic effects of PRM on metabolites are not clearly understood. Therefore, this study aimed to provide new information on the causal impact of PRM on the endogenous metabolites, pathways and serum biochemistry. Serum samples of hyperlipidemic rats treated with PRM were subjected to biochemistry (lipid and liver profile) and hydroxymethylglutaryl-CoA enzyme reductase (HMG-CoA reductase) analyses. In contrast, the urine samples were subjected to urine metabolomics using 1H NMR. The serum biochemistry revealed that PRM at 500 mg/kg (PRM-H) managed to lower the total cholesterol level and low-density lipoprotein (LDL-C) (p < 0.05) and reduce the HMG-CoA reductase activity. The pathway analysis from urine metabolomics reveals that PRM-H altered 17 pathways, with the TCA cycle having the highest impact (0.26). Results also showed the relationship between the serum biochemistry of LDL-C and HMG-CoA reductase and urine metabolites (trimethylamine-N-oxide, dimethylglycine, allantoin and succinate). The study's findings demonstrated the potential of PRM at 500 mg/kg as an anti-hyperlipidemic by altering the TCA cycle, inhibiting HMG-CoA reductase and lowering the LDL-C in high cholesterol rats.
Collapse
Affiliation(s)
- Azliana Abu Bakar Sajak
- Department of Nutrition, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia (UPM), Serdang 43400, Selangor, Malaysia;
| | - Azrina Azlan
- Department of Nutrition, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia (UPM), Serdang 43400, Selangor, Malaysia;
- Research Centre for Excellence for Nutrition and Non-Communicable Disease, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia (UPM), Serdang 43400, Selangor, Malaysia
| | - Faridah Abas
- Laboratory of Natural Products, Institute of Bioscience, Universiti Putra Malaysia (UPM), Serdang 43400, Selangor, Malaysia;
| | - Hazilawati Hamzah
- Department of Veterinary Medicine, Faculty of Veterinary Medicine, Universiti Putra Malaysia (UPM), Serdang 43400, Selangor, Malaysia;
| |
Collapse
|
119
|
Wang Y, Ning Y, Yuan C, Cui B, Liu G, Zhang Z. The protective mechanism of a debranched corn starch/konjac glucomannan composite against dyslipidemia and gut microbiota in high-fat-diet induced type 2 diabetes. Food Funct 2021; 12:9273-9285. [PMID: 34606538 DOI: 10.1039/d1fo01233a] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
This study aimed to explore the protection mechanism of a debranched corn starch/konjac glucomannan (DCSK) composite against type 2 diabetes (T2D) related to dyslipidemia and gut microbiota in mice fed on a high-fat diet (HFD). The results showed that the consumption of DCSK led to a significant improvement in the biochemical parameters and physiological indices associated with T2D in the HFD group, including the decrease in blood glucose, triglyceride, total cholesterol, and high-density lipoprotein cholesterol levels, as well as the suppression of the oxidative stress of the liver and kidneys. Furthermore, the health of the intestinal microbiota in the HFD-fed mice was altered dramatically after DCSK consumption. Metabolomics revealed 13 differential metabolites strongly linked to DCSK intervention, and DCSK supplementation regulated amino acid metabolism, nucleotide metabolism, and lipid metabolism. These findings demonstrated that DCSK has an outstanding ability to improve hyperglycemia, hyperlipidemia, and gut microbiota associated with T2D.
Collapse
Affiliation(s)
- Yanli Wang
- State Key Laboratory of Biobased Material and Green Papermaking, Qilu University of Technology, Shandong Academy of Sciences, Jinan, 250353, China. .,School of Grain and Oil, Henan University of Technology, Zhengzhou 450001, China
| | - Yuejia Ning
- State Key Laboratory of Biobased Material and Green Papermaking, Qilu University of Technology, Shandong Academy of Sciences, Jinan, 250353, China. .,School of Food Science and Engineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, China
| | - Chao Yuan
- State Key Laboratory of Biobased Material and Green Papermaking, Qilu University of Technology, Shandong Academy of Sciences, Jinan, 250353, China. .,School of Food Science and Engineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, China
| | - Bo Cui
- State Key Laboratory of Biobased Material and Green Papermaking, Qilu University of Technology, Shandong Academy of Sciences, Jinan, 250353, China. .,School of Food Science and Engineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, China
| | - Guimei Liu
- State Key Laboratory of Biobased Material and Green Papermaking, Qilu University of Technology, Shandong Academy of Sciences, Jinan, 250353, China. .,School of Food Science and Engineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, China
| | - Zheng Zhang
- State Key Laboratory of Biobased Material and Green Papermaking, Qilu University of Technology, Shandong Academy of Sciences, Jinan, 250353, China. .,School of Food Science and Engineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, China
| |
Collapse
|
120
|
Guo F, Si R, Li Q, Hai L, Yi L, He J, Ming L, Ji R. Reversible insulin resistance helps Bactrian camels survive fasting. Sci Rep 2021; 11:18815. [PMID: 34552154 PMCID: PMC8458433 DOI: 10.1038/s41598-021-98234-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 09/02/2021] [Indexed: 11/13/2022] Open
Abstract
Camels have hunger tolerance and can adapt to the severe environment of the desert. Through the comparison of insulin signalling pathway genes in different tissues in different eating periods (feeding, fasting and recovery feeding), it was found that IRS1, PIK3CB, PIK3R1 and SLC2A4 expression was significantly downregulated in the fore hump and hind hump during the fasting period. In addition, there was no difference in serum insulin levels among the three stages. However, the serum leptin and adiponectin levels decreased significantly during fasting. Additionally, insulin tolerance tests during the three stages showed that camels were insensitive to insulin during fasting. Further study of the serum metabolites showed that serum branched-chain and aromatic amino acid levels increased during the fasting period. Finally, analysis of microbial diversity in camel faeces at different stages showed that during the fasting period, the proportion of Firmicutes and Actinobacteria increased, while that of Bacteroides and the butyrate-producing bacterium Roseburia decreased. The results of this study show that fasting is accompanied by changes in the activation of insulin pathways in various camel tissues, normal insulin levels, and increased lipolysis and insulin resistance, which return to normal after eating.
Collapse
Affiliation(s)
- Fucheng Guo
- Key Laboratory of Dairy Biotechnology and Bioengineering, Ministry of Education, College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot, 010018, China
| | - Rendalai Si
- Key Laboratory of Dairy Biotechnology and Bioengineering, Ministry of Education, College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot, 010018, China.,Camel Research Institute of Inner Mongolia, Alxa, 737300, China
| | - Quanyun Li
- Key Laboratory of Dairy Biotechnology and Bioengineering, Ministry of Education, College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot, 010018, China
| | - Le Hai
- Key Laboratory of Dairy Biotechnology and Bioengineering, Ministry of Education, College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot, 010018, China
| | - Li Yi
- Key Laboratory of Dairy Biotechnology and Bioengineering, Ministry of Education, College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot, 010018, China
| | - Jing He
- Key Laboratory of Dairy Biotechnology and Bioengineering, Ministry of Education, College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot, 010018, China
| | - Liang Ming
- Key Laboratory of Dairy Biotechnology and Bioengineering, Ministry of Education, College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot, 010018, China.
| | - Rimutu Ji
- Key Laboratory of Dairy Biotechnology and Bioengineering, Ministry of Education, College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot, 010018, China. .,Camel Research Institute of Inner Mongolia, Alxa, 737300, China.
| |
Collapse
|
121
|
Parnell LD, Noel SE, Bhupathiraju SN, Smith CE, Haslam DE, Zhang X, Tucker KL, Ordovas JM, Lai CQ. Metabolite patterns link diet, obesity, and type 2 diabetes in a Hispanic population. Metabolomics 2021; 17:88. [PMID: 34553271 PMCID: PMC8458177 DOI: 10.1007/s11306-021-01835-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 09/01/2021] [Indexed: 01/04/2023]
Abstract
INTRODUCTION Obesity is a precursor of type 2 diabetes (T2D). OBJECTIVES Our aim was to identify metabolic signatures of T2D and dietary factors unique to obesity. METHODS We examined a subsample of the Boston Puerto Rican Health Study (BPRHS) population with a high prevalence of obesity and T2D at baseline (n = 806) and participants (without T2D at baseline) at 5-year follow-up (n = 412). We determined differences in metabolite profiles between T2D and non-T2D participants of the whole sample and according to abdominal obesity status. Enrichment analysis was performed to identify metabolic pathways that were over-represented by metabolites that differed between T2D and non-T2D participants. T2D-associated metabolites unique to obesity were examined for correlation with dietary food groups to understand metabolic links between dietary intake and T2D risk. False Discovery Rate method was used to correct for multiple testing. RESULTS Of 526 targeted metabolites, 179 differed between T2D and non-T2D in the whole sample, 64 in non-obese participants and 120 unique to participants with abdominal obesity. Twenty-four of 120 metabolites were replicated and were associated with T2D incidence at 5-year follow-up. Enrichment analysis pointed to three metabolic pathways that were overrepresented in obesity-associated T2D: phosphatidylethanolamine (PE), long-chain fatty acids, and glutamate metabolism. Elevated intakes of three food groups, energy-dense takeout food, dairy intake and sugar-sweetened beverages, associated with 13 metabolites represented by the three pathways. CONCLUSION Metabolic signatures of lipid and glutamate metabolism link obesity to T2D, in parallel with increased intake of dairy and sugar-sweetened beverages, thereby providing insight into the relationship between dietary habits and T2D risk.
Collapse
Affiliation(s)
- Laurence D Parnell
- USDA Agricultural Research Service, Nutrition and Genomics Laboratory, JM-USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA
| | - Sabrina E Noel
- Department of Biomedical and Nutritional Sciences, University of Massachusetts Lowell, Lowell, MA, USA
| | - Shilpa N Bhupathiraju
- Channing Division of Network Medicine, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA, USA
| | - Caren E Smith
- Nutrition and Genomics Laboratory, JM-USDA Human Nutrition Research Center On Aging at Tufts University, Boston, MA, USA
| | - Danielle E Haslam
- Channing Division of Network Medicine, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA, USA
| | - Xiyuang Zhang
- Department of Biomedical and Nutritional Sciences, University of Massachusetts Lowell, Lowell, MA, USA
| | - Katherine L Tucker
- Department of Biomedical and Nutritional Sciences, University of Massachusetts Lowell, Lowell, MA, USA
| | - Jose M Ordovas
- Nutrition and Genomics Laboratory, JM-USDA Human Nutrition Research Center On Aging at Tufts University, Boston, MA, USA
- IMDEA Food Institute, CEI UAM + CSIC, Madrid, Spain
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Chao-Qiang Lai
- USDA Agricultural Research Service, Nutrition and Genomics Laboratory, JM-USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA.
| |
Collapse
|
122
|
The Association between Branched-Chain Amino Acids (BCAAs) and Cardiometabolic Risk Factors in Middle-Aged Caucasian Women Stratified According to Glycemic Status. Nutrients 2021; 13:nu13103307. [PMID: 34684308 PMCID: PMC8538048 DOI: 10.3390/nu13103307] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 09/17/2021] [Accepted: 09/19/2021] [Indexed: 02/07/2023] Open
Abstract
We examined the glycemic status-stratified relationships between total serum branched-chain amino acid (BCAA) concentrations and cardiometabolic risk factors in middle-aged Caucasian women. The study included 349 women divided into 2 subgroups: a normoglycemic group (NG, n = 184) and a dysglycemic group (DG, n = 165). Blood samples, anthropometric parameters, and blood pressure were measured. HOMA-IR, albumin-corrected calcium (CCa), and fatty liver index (FLI) were calculated. BCAA concentrations were higher in the women with dysglycemia. BCAAs moderately correlated with BMI and FLI in the NG group and with BMI, FLI, total calcium (TCa), CCa, HbA1c, TG/HDL-C, and HDL-C in the DG group. After adjusting for age and BMI, correlations for TCa, CCa, HbA1c, HDL-C, and TG/HDL-C remained significant. The coexistence of increased BCAAs with dysglycemic status was associated with markedly higher concentrations of TCa, CCa, HbA1c, and TG, which were not observed in the DG women with low level of BCAAs. Multiple regression showed that TCa or CCa, age and BCAAs were significantly associated with HbA1c independently of BMI only in the DG group. We conclude that dysglycemia in particular predisposes women to a significant relationship between total BCAAs and circulating calcium and HbA1c, and that these relationships are independent of BMI and may reflect the pathophysiological calcium-dependent mechanisms connecting BCAAs with metabolic disturbances.
Collapse
|
123
|
Bozadjieva Kramer N, Evers SS, Shin JH, Silverwood S, Wang Y, Burant CF, Sandoval DA, Seeley RJ. The Role of Elevated Branched-Chain Amino Acids in the Effects of Vertical Sleeve Gastrectomy to Reduce Weight and Improve Glucose Regulation. Cell Rep 2021; 33:108239. [PMID: 33053352 PMCID: PMC7605275 DOI: 10.1016/j.celrep.2020.108239] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 06/24/2020] [Accepted: 09/15/2020] [Indexed: 12/16/2022] Open
Abstract
Elevated levels of branched-chain amino acids (BCAAs) and their metabolites are strongly positively associated with obesity, insulin resistance, and type 2 diabetes. Bariatric surgery is among the best treatments for weight loss and associated morbidities. Clinical studies have reported that bariatric surgery decreases the circulating levels of BCAAs. The objective of this study was to test the hypothesis that reduced BCAA levels contribute to the metabolic improvements of sustained weight loss and improved glucose tolerance after vertical sleeve gastrectomy (VSG). We find that, as in humans, circulating BCAAs are significantly lower in VSG rats and mice. To increase circulating BCAAs, we tested mice with either increased dietary intake of BCAAs or impaired BCAA catabolism by total body deletion of mitochondrial phosphatase 2C (Pp2cm). Our results show that a decrease in circulating BCAAs is not necessary for sustained body weight loss and improved glucose tolerance after VSG. Increased branched-chain amino acid (BCAA) levels are biomarkers of metabolic disease, and bariatric surgeries reduce BCAA levels. Bozadjieva Kramer et al. show that both dietary and genetic manipulations can block the surgical effect on BCAAs but do not alter potent, beneficial effects on weight loss and glucose tolerance.
Collapse
Affiliation(s)
| | - Simon S Evers
- Department of Surgery, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jae Hoon Shin
- Department of Surgery, University of Michigan, Ann Arbor, MI 48109, USA
| | - Sierra Silverwood
- Department of Surgery, University of Michigan, Ann Arbor, MI 48109, USA
| | - Yibin Wang
- Departments of Anesthesiology, Medicine, and Physiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Charles F Burant
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | | | - Randy J Seeley
- Department of Surgery, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
124
|
Qing Q, Huang L, Sun W, Chen J, Yu N, Chen Y, Xu D, Zhao M. Maternal and fetal metabolomic alterations in maternal lipopolysaccharide exposure-induced male offspring glucose metabolism disorders. Biomed Chromatogr 2021; 36:e5234. [PMID: 34477231 DOI: 10.1002/bmc.5234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 08/20/2021] [Accepted: 08/24/2021] [Indexed: 11/06/2022]
Abstract
Maternal lipopolysaccharide (LPS) exposure during pregnancy induces metabolic abnormalities in male offspring, but the underlying mechanisms remain unclear. The purpose of this study was to investigate the effects of maternal LPS exposure during pregnancy on metabolic profiling of maternal serum and male fetal liver using Liquid Chromatograph Mass Spectrometer techniques. From day 15 to day 17 of gestation, pregnant mice were administered intraperitoneal LPS (experimental group) (50 μg/kg/d) or saline (control group). On day 18 of gestation, maternal serum and male fetal liver were collected. After LPS exposure, levels of 38 and 75 metabolites, mainly glycerophospholipid and fatty acid metabolites, were altered in maternal serum and male fetal liver, respectively. It was found that in maternal serum and male fetal livers, the glycerophospholipids containing saturated fatty acids (SFAs) and the SFAs were upregulated, while the glycerophospholipids containing polyunsaturated fatty acids (PUFAs) and the PUFAs were downregulated. This concordance between maternal and fetal alterations in glycerophospholipid and fatty acid metabolites may be a metabolomic signature of the early intrauterine period and may provide insight into the mechanisms by which maternal LPS exposure induces disorders of glucose metabolism in male offspring.
Collapse
Affiliation(s)
- Qiting Qing
- School of Nursing, Anhui Medical University, Hefei, China
| | - Lili Huang
- School of Nursing, Anhui Medical University, Hefei, China
| | - Wanxiao Sun
- School of Nursing, Anhui Medical University, Hefei, China
| | - Jing Chen
- School of Nursing, Anhui Medical University, Hefei, China
| | - Ningning Yu
- School of Nursing, Anhui Medical University, Hefei, China
| | - Yuanhua Chen
- Department of Histology and Embryology, Anhui Medical University, Hefei, China
| | - Dexiang Xu
- Department of Toxicology, Anhui Medical University, Hefei, China
| | - Mei Zhao
- School of Nursing, Anhui Medical University, Hefei, China
| |
Collapse
|
125
|
Sjögren RJO, Rizo-Roca D, Chibalin AV, Chorell E, Furrer R, Katayama S, Harada J, Karlsson HKR, Handschin C, Moritz T, Krook A, Näslund E, Zierath JR. Branched-chain amino acid metabolism is regulated by ERRα in primary human myotubes and is further impaired by glucose loading in type 2 diabetes. Diabetologia 2021; 64:2077-2091. [PMID: 34131782 PMCID: PMC8382616 DOI: 10.1007/s00125-021-05481-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 03/12/2021] [Indexed: 11/26/2022]
Abstract
AIMS/HYPOTHESIS Increased levels of branched-chain amino acids (BCAAs) are associated with type 2 diabetes pathogenesis. However, most metabolomic studies are limited to an analysis of plasma metabolites under fasting conditions, rather than the dynamic shift in response to a metabolic challenge. Moreover, metabolomic profiles of peripheral tissues involved in glucose homeostasis are scarce and the transcriptomic regulation of genes involved in BCAA catabolism is partially unknown. This study aimed to identify differences in circulating and skeletal muscle BCAA levels in response to an OGTT in individuals with normal glucose tolerance (NGT) or type 2 diabetes. Additionally, transcription factors involved in the regulation of the BCAA gene set were identified. METHODS Plasma and vastus lateralis muscle biopsies were obtained from individuals with NGT or type 2 diabetes before and after an OGTT. Plasma and quadriceps muscles were harvested from skeletal muscle-specific Ppargc1a knockout and transgenic mice. BCAA-related metabolites and genes were assessed by LC-MS/MS and quantitative RT-PCR, respectively. Small interfering RNA and adenovirus-mediated overexpression techniques were used in primary human skeletal muscle cells to study the role of PPARGC1A and ESRRA in the expression of the BCAA gene set. Radiolabelled leucine was used to analyse the impact of oestrogen-related receptor α (ERRα) knockdown on leucine oxidation. RESULTS Impairments in BCAA catabolism in people with type 2 diabetes under fasting conditions were exacerbated after a glucose load. Branched-chain keto acids were reduced 37-56% after an OGTT in the NGT group, whereas no changes were detected in individuals with type 2 diabetes. These changes were concomitant with a stronger correlation with glucose homeostasis biomarkers and downregulated expression of branched-chain amino acid transaminase 2, branched-chain keto acid dehydrogenase complex subunits and 69% of downstream BCAA-related genes in skeletal muscle. In primary human myotubes overexpressing peroxisome proliferator-activated receptor γ coactivator-1α (PGC-1α, encoded by PPARGC1A), 61% of the analysed BCAA genes were upregulated, while 67% were downregulated in the quadriceps of skeletal muscle-specific Ppargc1a knockout mice. ESRRA (encoding ERRα) silencing completely abrogated the PGC-1α-induced upregulation of BCAA-related genes in primary human myotubes. CONCLUSIONS/INTERPRETATION Metabolic inflexibility in type 2 diabetes impacts BCAA homeostasis and attenuates the decrease in circulating and skeletal muscle BCAA-related metabolites after a glucose challenge. Transcriptional regulation of BCAA genes in primary human myotubes via PGC-1α is ERRα-dependent.
Collapse
Affiliation(s)
- Rasmus J O Sjögren
- Department of Molecular Medicine and Surgery, Integrative Physiology, Karolinska Institutet, Stockholm, Sweden
| | - David Rizo-Roca
- Department of Molecular Medicine and Surgery, Integrative Physiology, Karolinska Institutet, Stockholm, Sweden
| | - Alexander V Chibalin
- Department of Molecular Medicine and Surgery, Integrative Physiology, Karolinska Institutet, Stockholm, Sweden
| | - Elin Chorell
- Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | | | - Shintaro Katayama
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - Jun Harada
- Cardiovascular-Metabolics Research Laboratories, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - Håkan K R Karlsson
- Department of Molecular Medicine and Surgery, Integrative Physiology, Karolinska Institutet, Stockholm, Sweden
| | | | - Thomas Moritz
- Swedish Metabolomics Centre, Department of Forest Genetics and Plant Physiology, Swedish University of Agricultural Sciences, Umeå, Sweden
| | - Anna Krook
- Department of Physiology and Pharmacology, Integrative Physiology, Karolinska Institutet, Stockholm, Sweden
| | - Erik Näslund
- Division of Surgery, Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Juleen R Zierath
- Department of Molecular Medicine and Surgery, Integrative Physiology, Karolinska Institutet, Stockholm, Sweden.
- Department of Physiology and Pharmacology, Integrative Physiology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
126
|
Liu J, Wu S, Cheng Y, Liu Q, Su L, Yang Y, Zhang X, Wu M, Choi JI, Tong H. Sargassum fusiforme Alginate Relieves Hyperglycemia and Modulates Intestinal Microbiota and Metabolites in Type 2 Diabetic Mice. Nutrients 2021; 13:2887. [PMID: 34445047 PMCID: PMC8398017 DOI: 10.3390/nu13082887] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 08/17/2021] [Accepted: 08/19/2021] [Indexed: 12/15/2022] Open
Abstract
Sargassum fusiforme alginate (SF-Alg) possess many pharmacological activities, including hypoglycemic and hypolipidemic. However, the hypoglycemic mechanisms of SF-Alg remain unclear due to its low bioavailability. In this study, we evaluated the therapeutic effect of SF-Alg on high-fat diet (HFD)/streptozotocin (STZ)-induced type 2 diabetes (T2D) mice. SF-Alg intervention was found to significantly reduce fasting blood glucose (FBG), triglycerides (TG), and total cholesterol (TC), while increasing high-density lipoprotein cholesterol (HDL-c) and improving glucose tolerance. In addition, administrating SF-Alg to diabetic mice moderately attenuated pathological changes in adipose, hepatic, and heart tissues as well as skeletal muscle, and diminished oxidative stress. To probe the underlying mechanisms, we further analyzed the gut microbiota using 16S rRNA amplicon sequencing, as well as metabolites by non-targeted metabolomics. Here, SF-Alg significantly increased some benign bacteria (Lactobacillus, Bacteroides, Akkermansia Alloprevotella, Weissella and Enterorhabdus), and significantly decreased harmful bacteria (Turicibacter and Helicobacter). Meanwhile, SF-Alg dramatically decreased branched-chain amino acids (BCAAs) and aromatic amino acids (AAAs) in the colon of T2D mice, suggesting a positive benefit of SF-Alg as an adjvant agent for T2D.
Collapse
Affiliation(s)
- Jian Liu
- College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China; (J.L.); (S.W.); (Y.C.); (L.S.); (Y.Y.); (X.Z.); (M.W.)
- Department of Biotechnology and Bioengineering, Chonnam National University, Gwangju 500-757, Korea
| | - Siya Wu
- College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China; (J.L.); (S.W.); (Y.C.); (L.S.); (Y.Y.); (X.Z.); (M.W.)
| | - Yang Cheng
- College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China; (J.L.); (S.W.); (Y.C.); (L.S.); (Y.Y.); (X.Z.); (M.W.)
| | - Qiuhui Liu
- Bestchrom (Shanghai) Biosciences Co., Ltd., Shanghai 200120, China;
| | - Laijin Su
- College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China; (J.L.); (S.W.); (Y.C.); (L.S.); (Y.Y.); (X.Z.); (M.W.)
| | - Yue Yang
- College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China; (J.L.); (S.W.); (Y.C.); (L.S.); (Y.Y.); (X.Z.); (M.W.)
| | - Xu Zhang
- College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China; (J.L.); (S.W.); (Y.C.); (L.S.); (Y.Y.); (X.Z.); (M.W.)
| | - Mingjiang Wu
- College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China; (J.L.); (S.W.); (Y.C.); (L.S.); (Y.Y.); (X.Z.); (M.W.)
| | - Jong-il Choi
- Department of Biotechnology and Bioengineering, Chonnam National University, Gwangju 500-757, Korea
| | - Haibin Tong
- College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China; (J.L.); (S.W.); (Y.C.); (L.S.); (Y.Y.); (X.Z.); (M.W.)
| |
Collapse
|
127
|
A Metabolomic Analysis of the Sex-Dependent Hispanic Paradox. Metabolites 2021; 11:metabo11080552. [PMID: 34436492 PMCID: PMC8401672 DOI: 10.3390/metabo11080552] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 08/13/2021] [Accepted: 08/17/2021] [Indexed: 12/14/2022] Open
Abstract
In Mexican Americans, metabolic conditions, such as obesity and type 2 diabetes (T2DM), are not necessarily associated with an increase in mortality; this is the so-called Hispanic paradox. In this cross-sectional analysis, we used a metabolomic analysis to look at the mechanisms behind the Hispanic paradox. To do this, we examined dietary intake and body mass index (BMI; kg/m2) in men and women and their effects on serum metabolomic fingerprints in 70 Mexican Americans (26 men, 44 women). Although having different BMI values, the participants had many similar anthropometric and biochemical parameters, such as systolic and diastolic blood pressure, total cholesterol, and LDL cholesterol, which supported the paradox in these subjects. Plasma metabolomic phenotypes were measured using liquid chromatography tandem mass spectrometry (LC-MS/MS). A two-way ANOVA assessing sex, BMI, and the metabolome revealed 23 significant metabolites, such as 2-pyrrolidinone (p = 0.007), TMAO (p = 0.014), 2-aminoadipic acid (p = 0.019), and kynurenine (p = 0.032). Pathway and enrichment analyses discovered several significant metabolic pathways between men and women, including lysine degradation, tyrosine metabolism, and branch-chained amino acid (BCAA) degradation and biosynthesis. A log-transformed OPLS-DA model was employed and demonstrated a difference due to BMI in the metabolomes of both sexes. When stratified for caloric intake (<2200 kcal/d vs. >2200 kcal/d), a separate OPLS-DA model showed clear separation in men, while females remained relatively unchanged. After accounting for caloric intake and BMI status, the female metabolome showed substantial resistance to alteration. Therefore, we provide a better understanding of the Mexican-American metabolome, which may help demonstrate how this population—particularly women—possesses a longer life expectancy despite several comorbidities, and reveal the underlying mechanisms of the Hispanic paradox.
Collapse
|
128
|
Ning Z, Song Z, Wang C, Peng S, Wan X, Liu Z, Lu A. How Perturbated Metabolites in Diabetes Mellitus Affect the Pathogenesis of Hypertension? Front Physiol 2021; 12:705588. [PMID: 34483960 PMCID: PMC8416465 DOI: 10.3389/fphys.2021.705588] [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: 05/05/2021] [Accepted: 07/26/2021] [Indexed: 11/17/2022] Open
Abstract
The presence of hypertension (HTN) in type 2 diabetes mellitus (DM) is a common phenomenon in more than half of the diabetic patients. Since HTN constitutes a predictor of vascular complications and cardiovascular disease in type 2 DM patients, it is of significance to understand the molecular and cellular mechanisms of type 2 DM binding to HTN. This review attempts to understand the mechanism via the perspective of the metabolites. It reviewed the metabolic perturbations, the biological function of perturbated metabolites in two diseases, and the mechanism underlying metabolic perturbation that contributed to the connection of type 2 DM and HTN. DM-associated metabolic perturbations may be involved in the pathogenesis of HTN potentially in insulin, angiotensin II, sympathetic nervous system, and the energy reprogramming to address how perturbated metabolites in type 2 DM affect the pathogenesis of HTN. The recent integration of the metabolism field with microbiology and immunology may provide a wider perspective. Metabolism affects immune function and supports immune cell differentiation by the switch of energy. The diverse metabolites produced by bacteria modified the biological process in the inflammatory response of chronic metabolic diseases either. The rapidly evolving metabolomics has enabled to have a better understanding of the process of diseases, which is an important tool for providing some insight into the investigation of diseases mechanism. Metabolites served as direct modulators of biological processes were believed to assess the pathological mechanisms involved in diseases.
Collapse
Affiliation(s)
- Zhangchi Ning
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhiqian Song
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Chun Wang
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Shitao Peng
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiaoying Wan
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhenli Liu
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Aiping Lu
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| |
Collapse
|
129
|
Hu C, Jia W. Multi-omics profiling: the way towards precision medicine in metabolic diseases. J Mol Cell Biol 2021; 13:mjab051. [PMID: 34406397 PMCID: PMC8697344 DOI: 10.1093/jmcb/mjab051] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 06/19/2021] [Accepted: 06/21/2021] [Indexed: 12/12/2022] Open
Abstract
Metabolic diseases including type 2 diabetes mellitus (T2DM), non-alcoholic fatty liver disease (NAFLD), and metabolic syndrome (MetS) are alarming health burdens around the world, while therapies for these diseases are far from satisfying as their etiologies are not completely clear yet. T2DM, NAFLD, and MetS are all complex and multifactorial metabolic disorders based on the interactions between genetics and environment. Omics studies such as genetics, transcriptomics, epigenetics, proteomics, and metabolomics are all promising approaches in accurately characterizing these diseases. And the most effective treatments for individuals can be achieved via omics pathways, which is the theme of precision medicine. In this review, we summarized the multi-omics studies of T2DM, NAFLD, and MetS in recent years, provided a theoretical basis for their pathogenesis and the effective prevention and treatment, and highlighted the biomarkers and future strategies for precision medicine.
Collapse
Affiliation(s)
- Cheng Hu
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus,
Shanghai Clinical Center for Diabetes, Shanghai Jiao Tong University Affiliated Sixth
People's Hospital, Shanghai 200233, China
- Institute for Metabolic Disease, Fengxian Central Hospital, The Third School of
Clinical Medicine, Southern Medical University, Shanghai 201499, China
| | - Weiping Jia
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus,
Shanghai Clinical Center for Diabetes, Shanghai Jiao Tong University Affiliated Sixth
People's Hospital, Shanghai 200233, China
| |
Collapse
|
130
|
Wang QY, You LH, Xiang LL, Zhu YT, Zeng Y. Current progress in metabolomics of gestational diabetes mellitus. World J Diabetes 2021; 12:1164-1186. [PMID: 34512885 PMCID: PMC8394228 DOI: 10.4239/wjd.v12.i8.1164] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 05/20/2021] [Accepted: 07/07/2021] [Indexed: 02/06/2023] Open
Abstract
Gestational diabetes mellitus (GDM) is one of the most common metabolic disorders of pregnancy and can cause short- and long-term adverse effects in both pregnant women and their offspring. However, the etiology and pathogenesis of GDM are still unclear. As a metabolic disease, GDM is well suited to metabolomics study, which can monitor the changes in small molecular metabolites induced by maternal stimuli or perturbations in real time. The application of metabolomics in GDM can be used to discover diagnostic biomarkers, evaluate the prognosis of the disease, guide the application of diet or drugs, evaluate the curative effect, and explore the mechanism. This review provides comprehensive documentation of metabolomics research methods and techniques as well as the current progress in GDM research. We anticipate that the review will contribute to identifying gaps in the current knowledge or metabolomics technology, provide evidence-based information, and inform future research directions in GDM.
Collapse
Affiliation(s)
- Qian-Yi Wang
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 21000, Jiangsu Province, China
| | - Liang-Hui You
- Nanjing Maternity and Child Health Care Institute, Women’s Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing 21000, Jiangsu Province, China
| | - Lan-Lan Xiang
- Department of Clinical Laboratory, Women’s Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing 21000, Jiangsu Province, China
| | - Yi-Tian Zhu
- Department of Clinical Laboratory, Women’s Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing 21000, Jiangsu Province, China
| | - Yu Zeng
- Department of Clinical Laboratory, Women’s Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing 21000, Jiangsu Province, China
| |
Collapse
|
131
|
Xia F, Wen LP, Ge BC, Li YX, Li FP, Zhou BJ. Gut microbiota as a target for prevention and treatment of type 2 diabetes: Mechanisms and dietary natural products. World J Diabetes 2021; 12:1146-1163. [PMID: 34512884 PMCID: PMC8394227 DOI: 10.4239/wjd.v12.i8.1146] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 05/10/2021] [Accepted: 07/05/2021] [Indexed: 02/06/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM) is among the most remarkable public health concerns globally. Accumulating research evidence documents that alteration of gut microbiota has an indispensable role in the onset and progression of obesity and T2DM. A reduced microbial diversity is linked to insulin resistance and energy metabolism, especially for the rise of the Firmicutes/Bacteroidetes ratio. Changes in metabolites followed by the gut dysbacteriosis are linked to the presence of T2DM. Moreover, endotoxin leakage and gut permeability caused by gut dysbacteriosis is more of a trigger for the onset and progression of T2DM. Research documents that natural products are remarkable arsenals of bioactive agents for the discovery of anti-T2DM drugs. Many studies have elucidated that the possible mechanisms of the anti-T2DM effects of natural products are remarkably linked to its regulation on the composition of gut microflora and the successive changes in metabolites directly or indirectly. This review presents a brief overview of the gut microbiota in T2DM and several relevant mechanisms, including short-chain fatty acids, biosynthesis and metabolism of branched-chain fatty acids, trimethylamine N-oxide, bile acid signaling, endotoxin leakage, and gut permeability, and describes how dietary natural products can improve T2DM via the gut microbiota.
Collapse
Affiliation(s)
- Fan Xia
- Department of Pharmacy, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen 518107, Guangdong Province, China
| | - Lu-Ping Wen
- Department of Pharmacy, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen 518107, Guangdong Province, China
| | - Bing-Chen Ge
- Department of Pharmacy, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen 518107, Guangdong Province, China
| | - Yu-Xin Li
- Department of Pharmacology, Guangdong Medical University, Zhanjiang 524023, Guangdong Province, China
| | - Fang-Ping Li
- Department of Endocrinology, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen 518107, Guangdong Province, China
| | - Ben-Jie Zhou
- Department of Pharmacy, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen 518107, Guangdong Province, China
| |
Collapse
|
132
|
Taylor AL, Davis DE, Codreanu SG, Harrison FE, Sherrod SD, McLean JA. Targeted and Untargeted Mass Spectrometry Reveals the Impact of High-Fat Diet on Peripheral Amino Acid Regulation in a Mouse Model of Alzheimer's Disease. J Proteome Res 2021; 20:4405-4414. [PMID: 34382806 DOI: 10.1021/acs.jproteome.1c00344] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Recent research regarding amino acid metabolism has shown that there may be a link between obesity and Alzheimer's disease (AD). This work reports a metabolomics study using targeted and untargeted mass spectrometry-based metabolomic strategies to investigate this link. Targeted hydrophilic interaction liquid chromatography-triple quadrupole mass spectrometry and untargeted reversed-phase liquid chromatography-high resolution tandem mass spectrometry assays were developed to analyze the metabolic changes that occur in AD and obesity. APPSwe/PS1ΔE9 (APP/PSEN1) transgenic mice (to represent familial or early-onset AD) and wild-type littermate controls were fed either a high-fat diet (HFD, 60% kcal from lard) or a low-fat diet (LFD, 10% kcal from lard) from 2 months of age or a reversal diet (HFD, followed by LFD from 9.5 months). For targeted analyses, we applied the guidelines outlined in the Clinical and Laboratory Standards Institute (CLSI) LC-MS C62-A document and the U.S. Food and Drug Administration (FDA) bioanalytical method validation guidance for industry to evaluate the figures of merit of the assays. Our targeted and untargeted metabolomics results suggest that numerous peripheral pathways, specifically amino acid metabolism and fatty acid metabolism, were significantly affected by AD and diet. Multiple amino acids (including alanine, glutamic acid, leucine, isoleucine, and phenylalanine), carnitines, and members of the fatty acid oxidation pathway were significantly increased in APP/PSEN1 mice on HFD compared to those on LFD. More substantial effects and changes were observed in the APP/PSEN1 mice than in the WT mice, suggesting that they were more sensitive to an HFD. These dysregulated peripheral pathways include numerous amino acid pathways and fatty acid beta oxidation and suggest that obesity combined with AD further enhances cognitive impairment, possibly through aggravated mitochondrial dysfunction. Furthermore, partial reversibility of many altered pathways was observed, which highlights that diet change can mitigate the metabolic effects of AD. The same trends in individual amino acids were observed in both strategies, highlighting the biological validity of the results.
Collapse
Affiliation(s)
- Amelia L Taylor
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Don E Davis
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Simona G Codreanu
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37235, United States.,Center for Innovative Technology, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Fiona E Harrison
- Vanderbilt University Medical Center, Division of Diabetes, Endocrinology, and Metabolism, Vanderbilt University Medical Center, Nashville, Tennessee 37235, United States
| | - Stacy D Sherrod
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37235, United States.,Center for Innovative Technology, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - John A McLean
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37235, United States.,Center for Innovative Technology, Vanderbilt University, Nashville, Tennessee 37235, United States
| |
Collapse
|
133
|
Wu S, Zuo J, Cheng Y, Zhang Y, Zhang Z, Wu M, Yang Y, Tong H. Ethanol extract of Sargarsum fusiforme alleviates HFD/STZ-induced hyperglycemia in association with modulation of gut microbiota and intestinal metabolites in type 2 diabetic mice. Food Res Int 2021; 147:110550. [PMID: 34399527 DOI: 10.1016/j.foodres.2021.110550] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 06/03/2021] [Accepted: 06/16/2021] [Indexed: 01/16/2023]
Abstract
Type 2 diabetes mellitus (T2DM) is considered a rapidly growing chronic disease that threatens human health worldwide. Extracts of various seaweeds have been shown to have anti-diabetic activity. Sargarsum fusiforme, an edible brown seaweed, has been shown to possess anti-inflammatory, anti-diabetic and anti-obesity activities. In this study, we investigated the beneficial effect of an ethanol extract of S. fusiforme (EE) on type 2 diabetes in mice induced with high-fat diet (HFD) and streptozotocin (STZ). Administering EE to the diabetic mice significantly reduced food intake, water intake and fasting blood glucose (FBG), while improving glucose tolerance, lipid profile and ameliorating hepatic oxidative stress. Furthermore, these animals also exhibited significantly diminished epididymal fat deposition, as well as less pathological changes in the heart and liver tissues, while displaying some highly enriched benign gut bacteria (e.g., Intestinimonas, Oscillibacter, Lachnoclostridium, unidentified_Lachnospiraceae, Roseburia and Anaerotruncus) and a lower abundance of bacteria associated with diabetes or other metabolic diseases (e.g., Enterorhabdus and Romboutsia). Metabolomic analysis revealed reduced levels of branched-chain amino acids (BCAA), such as l-valine and l-isoleucine, aromatic amino acids (AAA), such as l-tyrosine and l-phenylalanine, and increased levels of 4-hydroxyphenylacetic acid (4-HPA) in the gut content, suggesting that EE may impact T2DM through modulation of these compounds in the gut of the animals. Taken together, the results implied that S. fusiforme may contain valuable active components other than polysaccharides that have potential benefit in alleviating T2DM.
Collapse
Affiliation(s)
- Siya Wu
- College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China
| | - Jihui Zuo
- College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China
| | - Yang Cheng
- College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China
| | - Ya Zhang
- College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China
| | - Zhongshan Zhang
- Key Laboratory of Vector Biology and Pathogen Control of Zhejiang Province, Huzhou University, Huzhou Cent Hosp, Huzhou 313000, China
| | - Mingjiang Wu
- College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China.
| | - Yue Yang
- College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China.
| | - Haibin Tong
- College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China.
| |
Collapse
|
134
|
Fang Z, Li L, Lu W, Zhao J, Zhang H, Lee YK, Chen W. Bifidobacterium affected the correlation between gut microbial composition, SCFA metabolism, and immunity in mice with DNFB-induced atopic dermatitis. FOOD BIOSCI 2021. [DOI: 10.1016/j.fbio.2020.100648] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
135
|
Alesi S, Ghelani D, Rassie K, Mousa A. Metabolomic Biomarkers in Gestational Diabetes Mellitus: A Review of the Evidence. Int J Mol Sci 2021; 22:ijms22115512. [PMID: 34073737 PMCID: PMC8197243 DOI: 10.3390/ijms22115512] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 05/20/2021] [Accepted: 05/20/2021] [Indexed: 12/14/2022] Open
Abstract
Gestational diabetes mellitus (GDM) is the fastest growing type of diabetes, affecting between 2 to 38% of pregnancies worldwide, varying considerably depending on diagnostic criteria used and sample population studied. Adverse obstetric outcomes include an increased risk of macrosomia, and higher rates of stillbirth, instrumental delivery, and birth trauma. Metabolomics, which is a platform used to analyse and characterise a large number of metabolites, is increasingly used to explore the pathophysiology of cardiometabolic conditions such as GDM. This review aims to summarise metabolomics studies in GDM (from inception to January 2021) in order to highlight prospective biomarkers for diagnosis, and to better understand the dysfunctional metabolic pathways underlying the condition. We found that the most commonly deranged pathways in GDM include amino acids (glutathione, alanine, valine, and serine), carbohydrates (2-hydroxybutyrate and 1,5-anhydroglucitol), and lipids (phosphatidylcholines and lysophosphatidylcholines). We also highlight the possibility of using certain metabolites as predictive markers for developing GDM, with the use of highly stratified modelling techniques. Limitations for metabolomic research are evaluated, and future directions for the field are suggested to aid in the integration of these findings into clinical practice.
Collapse
Affiliation(s)
- Simon Alesi
- Monash Centre for Health Research and Implementation (MCHRI), School of Public Health and Preventive Medicine, Monash University, Melbourne 3168, Australia; (S.A.); (D.G.); (K.R.)
| | - Drishti Ghelani
- Monash Centre for Health Research and Implementation (MCHRI), School of Public Health and Preventive Medicine, Monash University, Melbourne 3168, Australia; (S.A.); (D.G.); (K.R.)
| | - Kate Rassie
- Monash Centre for Health Research and Implementation (MCHRI), School of Public Health and Preventive Medicine, Monash University, Melbourne 3168, Australia; (S.A.); (D.G.); (K.R.)
- Department of Diabetes, Monash Health, Melbourne 3168, Australia
| | - Aya Mousa
- Monash Centre for Health Research and Implementation (MCHRI), School of Public Health and Preventive Medicine, Monash University, Melbourne 3168, Australia; (S.A.); (D.G.); (K.R.)
- Correspondence:
| |
Collapse
|
136
|
Zhong W, Deng Q, Deng X, Zhong Z, Hou J. Plasma Metabolomics of Acute Coronary Syndrome Patients Based on Untargeted Liquid Chromatography-Mass Spectrometry. Front Cardiovasc Med 2021; 8:616081. [PMID: 34095243 PMCID: PMC8172787 DOI: 10.3389/fcvm.2021.616081] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Accepted: 03/18/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Acute coronary syndrome (ACS) is the main cause of death and morbidity worldwide. The present study aims to investigate the altered metabolites in plasma from patients with ACS and sought to identify metabolic biomarkers for ACS. Methods: The plasma metabolomics profiles of 284 ACS patients and 130 controls were carried out based on an untargeted liquid chromatography coupled with tandem mass spectrometry (LC-MS) approach. Multivariate statistical methods, pathway enrichment analysis, and univariate receiver operating characteristic (ROC) curve analysis were performed. Results: A total of 328 and 194 features were determined in positive and negative electrospray ionization mode in the LC-MS analysis, respectively. Twenty-eight metabolites were found to be differentially expressed, in ACS patients relative to controls (p < 0.05). Pathway analysis revealed that these metabolites are mainly involved in synthesis and degradation of ketone bodies, phenylalanine metabolism, and arginine and proline metabolism. Furthermore, a diagnostic model was constructed based on the metabolites identified and the areas under the curve (AUC) for 5-oxo-D-proline, creatinine, phosphatidylethanolamine lyso 16:0, and LPC (20:4) range from 0.764 to 0.844. The higher AUC value of 0.905 was obtained for the combined detection of phosphatidylethanolamine lyso 16:0 and LPC (20:4). Conclusions: Differential metabolic profiles may be useful for the effective diagnosis of ACS and may provide additional insight into the molecular mechanisms underlying ACS.
Collapse
Affiliation(s)
- Wei Zhong
- Center for Cardiovascular Diseases, Meizhou People's Hospital (Huangtang Hospital), Meizhou Hospital Affiliated to Sun Yat-sen University, Meizhou, China.,Guangdong Provincial Engineering and Technology Research Center for Molecular Diagnostics of Cardiovascular Diseases, Meizhou, China.,Guangdong Provincial Key Laboratory of Precision Medicine and Clinical Translational Research of Hakka Population, Meizhou, China
| | - Qiaoting Deng
- Guangdong Provincial Engineering and Technology Research Center for Molecular Diagnostics of Cardiovascular Diseases, Meizhou, China.,Guangdong Provincial Key Laboratory of Precision Medicine and Clinical Translational Research of Hakka Population, Meizhou, China.,Research Experimental Center, Meizhou People's Hospital (Huangtang Hospital), Meizhou Hospital Affiliated to Sun Yat-sen University, Meizhou, China
| | - Xunwei Deng
- Guangdong Provincial Engineering and Technology Research Center for Molecular Diagnostics of Cardiovascular Diseases, Meizhou, China.,Guangdong Provincial Key Laboratory of Precision Medicine and Clinical Translational Research of Hakka Population, Meizhou, China.,Research Experimental Center, Meizhou People's Hospital (Huangtang Hospital), Meizhou Hospital Affiliated to Sun Yat-sen University, Meizhou, China
| | - Zhixiong Zhong
- Center for Cardiovascular Diseases, Meizhou People's Hospital (Huangtang Hospital), Meizhou Hospital Affiliated to Sun Yat-sen University, Meizhou, China.,Guangdong Provincial Engineering and Technology Research Center for Molecular Diagnostics of Cardiovascular Diseases, Meizhou, China.,Guangdong Provincial Key Laboratory of Precision Medicine and Clinical Translational Research of Hakka Population, Meizhou, China
| | - Jingyuan Hou
- Guangdong Provincial Engineering and Technology Research Center for Molecular Diagnostics of Cardiovascular Diseases, Meizhou, China.,Guangdong Provincial Key Laboratory of Precision Medicine and Clinical Translational Research of Hakka Population, Meizhou, China.,Research Experimental Center, Meizhou People's Hospital (Huangtang Hospital), Meizhou Hospital Affiliated to Sun Yat-sen University, Meizhou, China
| |
Collapse
|
137
|
Yoneshiro T, Kataoka N, Walejko JM, Ikeda K, Brown Z, Yoneshiro M, Crown SB, Osawa T, Sakai J, McGarrah RW, White PJ, Nakamura K, Kajimura S. Metabolic flexibility via mitochondrial BCAA carrier SLC25A44 is required for optimal fever. eLife 2021; 10:66865. [PMID: 33944778 PMCID: PMC8137140 DOI: 10.7554/elife.66865] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 05/02/2021] [Indexed: 01/21/2023] Open
Abstract
Importing necessary metabolites into the mitochondrial matrix is a crucial step of fuel choice during stress adaptation. Branched chain-amino acids (BCAAs) are essential amino acids needed for anabolic processes, but they are also imported into the mitochondria for catabolic reactions. What controls the distinct subcellular BCAA utilization during stress adaptation is insufficiently understood. The present study reports the role of SLC25A44, a recently identified mitochondrial BCAA carrier (MBC), in the regulation of mitochondrial BCAA catabolism and adaptive response to fever in rodents. We found that mitochondrial BCAA oxidation in brown adipose tissue (BAT) is significantly enhanced during fever in response to the pyrogenic mediator prostaglandin E2 (PGE2) and psychological stress in mice and rats. Genetic deletion of MBC in a BAT-specific manner blunts mitochondrial BCAA oxidation and non-shivering thermogenesis following intracerebroventricular PGE2 administration. At a cellular level, MBC is required for mitochondrial BCAA deamination as well as the synthesis of mitochondrial amino acids and TCA intermediates. Together, these results illuminate the role of MBC as a determinant of metabolic flexibility to mitochondrial BCAA catabolism and optimal febrile responses. This study also offers an opportunity to control fever by rewiring the subcellular BCAA fate.
Collapse
Affiliation(s)
- Takeshi Yoneshiro
- Diabetes Center and Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, United States.,Division of Metabolic Medicine, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Naoya Kataoka
- Department of Integrative Physiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Jacquelyn M Walejko
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, United States
| | - Kenji Ikeda
- Diabetes Center and Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, United States.,Department of Molecular Endocrinology and Metabolism, Tokyo Medical and Dental University, Tokyo, Japan
| | - Zachary Brown
- Diabetes Center and Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, United States
| | - Momoko Yoneshiro
- Diabetes Center and Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, United States
| | - Scott B Crown
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, United States
| | - Tsuyoshi Osawa
- Division of Integrative Nutriomics and Oncology, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Juro Sakai
- 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
| | - Robert W McGarrah
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, United States.,Department of Medicine, Division of Cardiology, Duke University School of Medicine, Durham, United States
| | - Phillip J White
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, United States.,Department of Medicine, Division of EndocrinologyMetabolism and Nutrition, Duke University School of Medicine, Durham, United States
| | - Kazuhiro Nakamura
- Department of Integrative Physiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shingo Kajimura
- Diabetes Center and Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, United States.,Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Durham, United States
| |
Collapse
|
138
|
The effect of physical activity level and exercise training on the association between plasma branched-chain amino acids and intrahepatic lipid content in participants with obesity. Int J Obes (Lond) 2021; 45:1510-1520. [PMID: 33935282 PMCID: PMC8236400 DOI: 10.1038/s41366-021-00815-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 02/01/2021] [Accepted: 04/09/2021] [Indexed: 01/05/2023]
Abstract
Aims To evaluate whether the association between plasma branched-chain amino acids (BCAA) and intrahepatic lipid (IHL) was affected by physical activity level. Furthermore, to investigate if a conventional exercise training program, a subcategory of physical activity, could lower plasma BCAA along with alterations in IHL content in patients with type 2 diabetes (T2DM) and people with nonalcoholic fatty liver (NAFL). Methods To investigate the effect of physical activity on the association between plasma BCAA and IHL content, linear regression analyses were performed in 1983 individuals from the Netherlands Epidemiology of Obesity (NEO) stratified by physical activity frequency. Furthermore, the effect of a 12-week supervised combined aerobic resistance-exercise program on plasma BCAA, insulin sensitivity (hyperinsulinemic–euglycemic clamp), and IHL (proton-magnetic resonance spectroscopy (1H-MRS)) was investigated in seven patients with T2DM, seven individuals with NAFL and seven BMI-matched control participants (CON). Results We observed positive associations between plasma valine, isoleucine and leucine level, and IHL content (1.29 (95% CI: 1.21, 1.38), 1.52 (95% CI: 1.43, 1.61), and 1.54 (95% CI: 1.44, 1.64) times IHL, respectively, per standard deviation of plasma amino acid level). Similar associations were observed in less active versus more active individuals. Exercise training did not change plasma BCAA levels among groups, but reduced IHL content in NAFL (from 11.6 ± 3.0% pre-exercise to 8.1 ± 2.0% post exercise, p < 0.05) and CON (from 2.4 ± 0.6% pre-exercise to 1.6 ± 1.4% post exercise, p < 0.05), and improved peripheral insulin sensitivity in NAFL as well by ~23% (p < 0.05). Conclusions The association between plasma BCAA levels and IHL is not affected by physical activity level. Exercise training reduced IHL without affecting plasma BCAA levels in individuals with NAFL and CON. We conclude that exercise training-induced reduction in IHL content is not related to changes in plasma BCAA levels. Trial registration Trial registry number: NCT01317576.
Collapse
|
139
|
Amino Acid Signature of Oxidative Stress in Patients with Type 2 Diabetes: Targeted Exploratory Metabolomic Research. Antioxidants (Basel) 2021; 10:antiox10040610. [PMID: 33921149 PMCID: PMC8071553 DOI: 10.3390/antiox10040610] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 04/11/2021] [Accepted: 04/12/2021] [Indexed: 02/06/2023] Open
Abstract
Oxidative stress plays a key role in the development of chronic diabetes-related complications. Previous metabolomic studies showed a positive association of diabetes and insulin resistance with branched-chain amino acids (AAs) and aromatic AAs. The purpose of this research is to identify distinct metabolic changes associated with increased oxidative stress, as assessed by nitrotyrosine levels, in type 2 diabetes (T2DM). Serum samples of 80 patients with insulin-treated T2DM are analyzed by AA-targeted metabolomics using ultrahigh-performance liquid chromatography/mass spectrometry. Patients are divided into two groups based on their nitrotyrosine levels: the highest level of oxidative stress (Q4 nitrotyrosine) and lower levels (Q1–Q3 nitrotyrosine). The identification of biomarkers is performed in MetaboAnalyst version 5.0 using a t-test corrected for false discovery rate, unsupervised principal component analysis and supervised partial least-squares discriminant analysis (PLS-DA). Four AAs have significantly different levels between the groups for highest and lower oxidative stress. Cysteine, phenylalanine and tyrosine are substantially increased while citrulline is decreased (p-value <0.05 and variable importance in the projection [VIP] >1). Corresponding pathways that might be disrupted in patients with high oxidative stress are phenylalanine, tyrosine and tryptophan biosynthesis, arginine biosynthesis, phenylalanine metabolism, cysteine and methionine metabolism and tyrosine metabolism.
Collapse
|
140
|
Wu J, Zhao M, Li C, Zhang Y, Wang DW. The SARS-CoV-2 induced targeted amino acid profiling in patients at hospitalized and convalescent stage. Biosci Rep 2021; 41:BSR20204201. [PMID: 33625490 PMCID: PMC7955102 DOI: 10.1042/bsr20204201] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Revised: 02/10/2021] [Accepted: 02/23/2021] [Indexed: 02/07/2023] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has induced an ongoing global health crisis. Here we utilized a combination of targeted amino acids (AAs) and clinical biochemical profiling to analyze the plasma of coronavirus disease 2019 (COVID-19) subjects at the hospitalization stage and 1-month post-infection convalescent stage, respectively, to investigate the systematic injury during COVID-19 disease progress. We found the virus-induced inflammatory status and reduced liver synthesis capacity in hospitalized patients, which manifested with increased branched-chain AAs (BCAAs), aromatic AAs (AAAs), one-carbon related metabolites, and decreased methionine. Most of these disturbances during infection recover except for the increased levels of medium-chain acylcarnitines (ACs) in the convalescent subjects, implying the existence of incomplete fatty acids oxidation during recovery periods. Our results suggested that the imbalance of the AA profiling in COVID-19 patients. The majority of disturbed AAs recovered in 1 month. The incomplete fatty acid oxidation products suggested it might take longer time for convalescent patients to get complete recovery.
Collapse
Affiliation(s)
- Junfang Wu
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China
| | - Mingming Zhao
- The Institute of Cardiovascular Sciences and Institute of Systems Biomedicine, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Sciences of Ministry of Education, Health Science Center, Peking University, Beijing 100191, China
| | - Chenze Li
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China
| | - Yuxuan Zhang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China
| | - Dao Wen Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China
| |
Collapse
|
141
|
Vanweert F, de Ligt M, Hoeks J, Hesselink MKC, Schrauwen P, Phielix E. Elevated Plasma Branched-Chain Amino Acid Levels Correlate With Type 2 Diabetes-Related Metabolic Disturbances. J Clin Endocrinol Metab 2021; 106:e1827-e1836. [PMID: 33079174 DOI: 10.1210/clinem/dgaa751] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Accepted: 10/14/2020] [Indexed: 01/14/2023]
Abstract
CONTEXT Patients with type 2 diabetes mellitus (T2DM) have elevated plasma branched-chain amino acid (BCAA) levels. The underlying cause, however, is not known. Low mitochondrial oxidation of BCAA levels could contribute to higher plasma BCAA levels. OBJECTIVE We aimed to investigate ex vivo muscle mitochondrial oxidative capacity and in vivo BCAA oxidation measured by whole-body leucine oxidation rates in patients with T2DM, first-degree relatives (FDRs), and control participants (CONs) with overweight or obesity. DESIGN AND SETTING An observational, community-based study was conducted. PARTICIPANTS Fifteen patients with T2DM, 13 FDR, and 17 CONs were included (age, 40-70 years; body mass index, 27-35 kg/m2). MAIN OUTCOME MEASURES High-resolution respirometry was used to examine ex vivo mitochondrial oxidative capacity in permeabilized muscle fibers. A subgroup of 5 T2DM patients and 5 CONs underwent hyperinsulinemic-euglycemic clamps combined with 1-13C leucine-infusion to determine whole-body leucine oxidation. RESULTS Total BCAA levels were higher in patients with T2DM compared to CONs, but not in FDRs, and correlated negatively with muscle mitochondrial oxidative capacity (r = -0.44, P < .001). Consistently, whole-body leucine oxidation rate was lower in patients with T2DM vs CON under basal conditions (0.202 ± 0.049 vs 0.275 ± 0.043 μmol kg-1 min-1, P < .05) and tended to be lower during high insulin infusion (0.326 ± 0.024 vs 0.382 ± 0.013 μmol kg-1 min-1, P = .075). CONCLUSIONS In patients with T2DM, a compromised whole-body leucine oxidation rate supports our hypothesis that higher plasma BCAA levels may originate at least partly from a low mitochondrial oxidative capacity.
Collapse
Affiliation(s)
- Froukje Vanweert
- Department of Nutrition and Movement Sciences, Maastricht University and Medical Center, Maastricht, the Netherlands
- NUTRIM, School of Nutrition and Translational Research in Metabolism, Maastricht University and Medical Center, Maastricht, the Netherlands
| | - Marlies de Ligt
- Department of Nutrition and Movement Sciences, Maastricht University and Medical Center, Maastricht, the Netherlands
- NUTRIM, School of Nutrition and Translational Research in Metabolism, Maastricht University and Medical Center, Maastricht, the Netherlands
| | - Joris Hoeks
- Department of Nutrition and Movement Sciences, Maastricht University and Medical Center, Maastricht, the Netherlands
- NUTRIM, School of Nutrition and Translational Research in Metabolism, Maastricht University and Medical Center, Maastricht, the Netherlands
| | - Matthijs K C Hesselink
- Department of Nutrition and Movement Sciences, Maastricht University and Medical Center, Maastricht, the Netherlands
- NUTRIM, School of Nutrition and Translational Research in Metabolism, Maastricht University and Medical Center, Maastricht, the Netherlands
| | - Patrick Schrauwen
- Department of Nutrition and Movement Sciences, Maastricht University and Medical Center, Maastricht, the Netherlands
- NUTRIM, School of Nutrition and Translational Research in Metabolism, Maastricht University and Medical Center, Maastricht, the Netherlands
| | - Esther Phielix
- Department of Nutrition and Movement Sciences, Maastricht University and Medical Center, Maastricht, the Netherlands
- NUTRIM, School of Nutrition and Translational Research in Metabolism, Maastricht University and Medical Center, Maastricht, the Netherlands
| |
Collapse
|
142
|
Dietary Management of Type 2 Diabetes in the MENA Region: A Review of the Evidence. Nutrients 2021; 13:nu13041060. [PMID: 33805161 PMCID: PMC8064070 DOI: 10.3390/nu13041060] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 03/21/2021] [Accepted: 03/21/2021] [Indexed: 12/11/2022] Open
Abstract
The alarmingly rising trend of type 2 diabetes constitutes a major global public health challenge particularly in the Middle Eastern and North African (MENA) region where the prevalence is among the highest in the world with a projection to increase by 96% by 2045. The economic boom in the MENA region over the past decades has brought exceptionally rapid shifts in eating habits characterized by divergence from the traditional Mediterranean diet towards a more westernized unhealthy dietary pattern, thought to be leading to the dramatic rises in obesity and non-communicable diseases. Research efforts have brought a greater understanding of the different pathways through which diet and obesity may affect diabetes clinical outcomes, emphasizing the crucial role of dietary interventions and weight loss in the prevention and management of diabetes. The purpose of this review is to explore the mechanistic pathways linking obesity with diabetes and to summarize the most recent evidence on the association of the intake of different macronutrients and food groups with the risk of type 2 diabetes. We also summarize the most recent evidence on the effectiveness of different macronutrient manipulations in the prevention and management of diabetes while highlighting the possible underlying mechanisms of action and latest evidence-based recommendations. We finally discuss the need to adequately integrate dietetic services in diabetes care specific to the MENA region and conclude with recommendations to improve dietetic care for diabetes in the region.
Collapse
|
143
|
Lécuyer L, Victor Bala A, Demidem A, Rossary A, Bouchemal N, Triba MN, Galan P, Hercberg S, Partula V, Srour B, Latino-Martel P, Kesse-Guyot E, Druesne-Pecollo N, Vasson MP, Deschasaux-Tanguy M, Savarin P, Touvier M. NMR metabolomic profiles associated with long-term risk of prostate cancer. Metabolomics 2021; 17:32. [PMID: 33704614 DOI: 10.1007/s11306-021-01780-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 02/24/2021] [Indexed: 12/25/2022]
Abstract
INTRODUCTION Prostate cancer is a multifactorial disease whose aetiology is still not fully understood. Metabolomics, by measuring several hundred metabolites simultaneously, could enhance knowledge on the metabolic changes involved and the potential impact of external factors. OBJECTIVES The aim of the present study was to investigate whether pre-diagnostic plasma metabolomic profiles were associated with the risk of developing a prostate cancer within the following decade. METHODS A prospective nested case-control study was set up among the 5141 men participant of the SU.VI.MAX cohort, including 171 prostate cancer cases, diagnosed between 1994 and 2007, and 171 matched controls. Nuclear magnetic resonance (NMR) metabolomic profiles were established from baseline plasma samples using NOESY1D and CPMG sequences. Multivariable conditional logistic regression models were computed for each individual NMR signal and for metabolomic patterns derived using principal component analysis. RESULTS Men with higher fasting plasma levels of valine (odds ratio (OR) = 1.37 [1.07-1.76], p = .01), glutamine (OR = 1.30 [1.00-1.70], p = .047), creatine (OR = 1.37 [1.04-1.80], p = .02), albumin lysyl (OR = 1.48 [1.12-1.95], p = .006 and OR = 1.51 [1.13-2.02], p = .005), tyrosine (OR = 1.40 [1.06-1.85], p = .02), phenylalanine (OR = 1.39 [1.08-1.79], p = .01), histidine (OR = 1.46 [1.12-1.88], p = .004), 3-methylhistidine (OR = 1.37 [1.05-1.80], p = .02) and lower plasma level of urea (OR = .70 [.54-.92], p = .009) had a higher risk of developing a prostate cancer during the 13 years of follow-up. CONCLUSIONS This exploratory study highlighted associations between baseline plasma metabolomic profiles and long-term risk of developing prostate cancer. If replicated in independent cohort studies, such signatures may improve the identification of men at risk for prostate cancer well before diagnosis and the understanding of this disease.
Collapse
Affiliation(s)
- Lucie Lécuyer
- Inserm U1153, Inrae U1125, Cnam, Nutritional Epidemiology Research Team (EREN), Epidemiology and Statistics Research Center - University of Paris (CRESS), Sorbonne Paris Nord University, SMBH Paris 13, 74 rue Marcel Cachin, 93017, Bobigny Cedex, France
| | - Agnès Victor Bala
- Chemistry Structures Properties of Biomaterials and Therapeutic Agents (CSPBAT), Nanomédecine Biomarqueurs Détection (NBD), The National Center for Scientific Research (CNRS) 7244, Sorbonne Paris Nord University, 93017, Bobigny Cedex, France
| | - Aicha Demidem
- INRAE, UMR 1019, Human Nutrition Unit (UNH), Cellular Micro-Environment, Immunomodulation and Nutrition (ECREIN), Clermont Auvergne University, CRNH Auvergne, 63000, Clermont-Ferrand, France
| | - Adrien Rossary
- INRAE, UMR 1019, Human Nutrition Unit (UNH), Cellular Micro-Environment, Immunomodulation and Nutrition (ECREIN), Clermont Auvergne University, CRNH Auvergne, 63000, Clermont-Ferrand, France
| | - Nadia Bouchemal
- Chemistry Structures Properties of Biomaterials and Therapeutic Agents (CSPBAT), Nanomédecine Biomarqueurs Détection (NBD), The National Center for Scientific Research (CNRS) 7244, Sorbonne Paris Nord University, 93017, Bobigny Cedex, France
| | - Mohamed Nawfal Triba
- Chemistry Structures Properties of Biomaterials and Therapeutic Agents (CSPBAT), Nanomédecine Biomarqueurs Détection (NBD), The National Center for Scientific Research (CNRS) 7244, Sorbonne Paris Nord University, 93017, Bobigny Cedex, France
| | - Pilar Galan
- Inserm U1153, Inrae U1125, Cnam, Nutritional Epidemiology Research Team (EREN), Epidemiology and Statistics Research Center - University of Paris (CRESS), Sorbonne Paris Nord University, SMBH Paris 13, 74 rue Marcel Cachin, 93017, Bobigny Cedex, France
| | - Serge Hercberg
- Inserm U1153, Inrae U1125, Cnam, Nutritional Epidemiology Research Team (EREN), Epidemiology and Statistics Research Center - University of Paris (CRESS), Sorbonne Paris Nord University, SMBH Paris 13, 74 rue Marcel Cachin, 93017, Bobigny Cedex, France
- Public Health Department, Avicenne Hospital, 93000, Bobigny, France
| | - Valentin Partula
- Inserm U1153, Inrae U1125, Cnam, Nutritional Epidemiology Research Team (EREN), Epidemiology and Statistics Research Center - University of Paris (CRESS), Sorbonne Paris Nord University, SMBH Paris 13, 74 rue Marcel Cachin, 93017, Bobigny Cedex, France
| | - Bernard Srour
- Inserm U1153, Inrae U1125, Cnam, Nutritional Epidemiology Research Team (EREN), Epidemiology and Statistics Research Center - University of Paris (CRESS), Sorbonne Paris Nord University, SMBH Paris 13, 74 rue Marcel Cachin, 93017, Bobigny Cedex, France
| | - Paule Latino-Martel
- Inserm U1153, Inrae U1125, Cnam, Nutritional Epidemiology Research Team (EREN), Epidemiology and Statistics Research Center - University of Paris (CRESS), Sorbonne Paris Nord University, SMBH Paris 13, 74 rue Marcel Cachin, 93017, Bobigny Cedex, France
| | - Emmanuelle Kesse-Guyot
- Inserm U1153, Inrae U1125, Cnam, Nutritional Epidemiology Research Team (EREN), Epidemiology and Statistics Research Center - University of Paris (CRESS), Sorbonne Paris Nord University, SMBH Paris 13, 74 rue Marcel Cachin, 93017, Bobigny Cedex, France
| | - Nathalie Druesne-Pecollo
- Inserm U1153, Inrae U1125, Cnam, Nutritional Epidemiology Research Team (EREN), Epidemiology and Statistics Research Center - University of Paris (CRESS), Sorbonne Paris Nord University, SMBH Paris 13, 74 rue Marcel Cachin, 93017, Bobigny Cedex, France
| | - Marie-Paule Vasson
- INRAE, UMR 1019, Human Nutrition Unit (UNH), Cellular Micro-Environment, Immunomodulation and Nutrition (ECREIN), Clermont Auvergne University, CRNH Auvergne, 63000, Clermont-Ferrand, France
- Anticancer Center Jean-Perrin, CHU Clermont-Ferrand, 63011, Clermont-Ferrand Cedex, France
| | - Mélanie Deschasaux-Tanguy
- Inserm U1153, Inrae U1125, Cnam, Nutritional Epidemiology Research Team (EREN), Epidemiology and Statistics Research Center - University of Paris (CRESS), Sorbonne Paris Nord University, SMBH Paris 13, 74 rue Marcel Cachin, 93017, Bobigny Cedex, France.
| | - Philippe Savarin
- Chemistry Structures Properties of Biomaterials and Therapeutic Agents (CSPBAT), Nanomédecine Biomarqueurs Détection (NBD), The National Center for Scientific Research (CNRS) 7244, Sorbonne Paris Nord University, 93017, Bobigny Cedex, France
| | - Mathilde Touvier
- Inserm U1153, Inrae U1125, Cnam, Nutritional Epidemiology Research Team (EREN), Epidemiology and Statistics Research Center - University of Paris (CRESS), Sorbonne Paris Nord University, SMBH Paris 13, 74 rue Marcel Cachin, 93017, Bobigny Cedex, France
| |
Collapse
|
144
|
Whole-body metabolic fate of branched-chain amino acids. Biochem J 2021; 478:765-776. [PMID: 33626142 DOI: 10.1042/bcj20200686] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 02/01/2021] [Accepted: 02/03/2021] [Indexed: 12/12/2022]
Abstract
Oxidation of branched-chain amino acids (BCAAs) is tightly regulated in mammals. We review here the distribution and regulation of whole-body BCAA oxidation. Phosphorylation and dephosphorylation of the rate-limiting enzyme, branched-chain α-ketoacid dehydrogenase complex directly regulates BCAA oxidation, and various other indirect mechanisms of regulation also exist. Most tissues throughout the body are capable of BCAA oxidation, and the flux of oxidative BCAA disposal in each tissue is influenced by three key factors: 1. tissue-specific preference for BCAA oxidation relative to other fuels, 2. the overall oxidative activity of mitochondria within a tissue, and 3. total tissue mass. Perturbations in BCAA oxidation have been implicated in many disease contexts, underscoring the importance of BCAA homeostasis in overall health.
Collapse
|
145
|
Virgiliou C, Theodoridis G, Wilson ID, Gika HG. Quantification of endogenous aminoacids and aminoacid derivatives in urine by hydrophilic interaction liquid chromatography tandem mass spectrometry. J Chromatogr A 2021; 1642:462005. [PMID: 33657487 DOI: 10.1016/j.chroma.2021.462005] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 01/14/2021] [Accepted: 02/15/2021] [Indexed: 11/17/2022]
Abstract
Aminoacids and their derivatives are key biologically important metabolites and reliable, rapid and accurate, quantification for these analytes in urine remains an important analytical challenge. Here a fast and reliable HILIC-tandem MS method is presented for application in clinical or nutritional studies. The developed method was validated according to existing guidelines adapted for endogenous analytes. The validation strategy provided evidence of linearity, LOD and LOQ, accuracy, precision, matrix effect and recovery. The surrogate matrix approach was applied for calibration proving satisfactory accuracy and precision based on standard criteria over the working concentration ranges. Intra and inter day accuracy was found to range between 0.8 and 20% for the LQC (low QC) and between 0.05 and 15 % for MQC (medium QC) and HQC (high QC). Inter and intraday precision were found to be between 3 and 20 % for the LQC and between 1 and 15% for the MQC and HQC. The stability of the analytes, in both surrogate and pooled urine QC samples, was found to be within 15% over a short period at 4 °C or after a up to 3 freeze-thaw cycles. The uncertainty of the method was also assessed to provide increased confidence for the acquired measurements. The method was successfully applied to a subset of human urine samples involved in a study of amino acids dietary uptake. This method may provide a valuable tool for many applications or studies where amino acid metabolic signatures in the excreted urine are under investigation.
Collapse
Affiliation(s)
- Christina Virgiliou
- Department of Chemistry, Aristotle University of Thessaloniki, 54124, Greece; Biomic_Auth, Bioanalysis and Omics Lab, CIRI - AUTH, Center for Interdisciplinary Research and Innovation, Thermi, 57001, Greece
| | - Georgios Theodoridis
- Department of Chemistry, Aristotle University of Thessaloniki, 54124, Greece; Biomic_Auth, Bioanalysis and Omics Lab, CIRI - AUTH, Center for Interdisciplinary Research and Innovation, Thermi, 57001, Greece
| | - Ian D Wilson
- Division of Systems Medicine, Department of Metabolism Department of Metabolism, Digestion and Reproduction, Imperial College London, South Kensigton, London SW7 2AZ, UK
| | - Helen G Gika
- Biomic_Auth, Bioanalysis and Omics Lab, CIRI - AUTH, Center for Interdisciplinary Research and Innovation, Thermi, 57001, Greece; School of Medicine, Aristotle University of Thessaloniki, 54124, Greece.
| |
Collapse
|
146
|
Lehtovirta M, Matthews LA, Laitinen TT, Nuotio J, Niinikoski H, Rovio SP, Lagström H, Viikari JSA, Rönnemaa T, Jula A, Ala-Korpela M, Raitakari OT, Pahkala K. Achievement of the Targets of the 20-Year Infancy-Onset Dietary Intervention-Association with Metabolic Profile from Childhood to Adulthood. Nutrients 2021; 13:nu13020533. [PMID: 33562015 PMCID: PMC7915301 DOI: 10.3390/nu13020533] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 01/28/2021] [Accepted: 02/02/2021] [Indexed: 02/06/2023] Open
Abstract
The Special Turku Coronary Risk Factor Intervention Project (STRIP) is a prospective infancy-onset randomized dietary intervention trial targeting dietary fat quality and cholesterol intake, and favoring consumption of vegetables, fruit, and whole-grains. Diet (food records) and circulating metabolites were studied at six time points between the ages of 9-19 years (n = 549-338). Dietary targets for this study were defined as (1) the ratio of saturated fat (SAFA) to monounsaturated and polyunsaturated fatty acids (MUFA + PUFA) < 1:2, (2) intake of SAFA < 10% of total energy intake, (3) fiber intake ≥ 80th age-specific percentile, and (4) sucrose intake ≤ 20th age-specific percentile. Metabolic biomarkers were quantified by high-throughput nuclear magnetic resonance metabolomics. Better adherence to the dietary targets, regardless of study group allocation, was assoiated with higher serum proportion of PUFAs, lower serum proportion of SAFAs, and a higher degree of unsaturation of fatty acids. Achieving ≥ 1 dietary target resulted in higher low-density lipoprotein (LDL) particle size, lower circulating LDL subclass lipid concentrations, and lower circulating lipid concentrations in medium and small high-density lipoprotein subclasses compared to meeting 0 targets. Attaining more dietary targets (≥2) was associated with a tendency to lower lipid concentrations of intermediate-density lipoprotein and very low-density lipoprotein subclasses. Thus, adherence to dietary targets is favorably associated with multiple circulating fatty acids and lipoprotein subclass lipid concentrations, indicative of better cardio-metabolic health.
Collapse
Affiliation(s)
- Miia Lehtovirta
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, 20520 Turku, Finland; (L.A.M.); (T.T.L.); (J.N.); (S.P.R.); (O.T.R.); (K.P.)
- Centre for Population Health Research, Turku University Hospital, University of Turku, 20520 Turku, Finland;
- Correspondence: ; Tel.: +358-2333-7552
| | - Laurie A. Matthews
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, 20520 Turku, Finland; (L.A.M.); (T.T.L.); (J.N.); (S.P.R.); (O.T.R.); (K.P.)
- Centre for Population Health Research, Turku University Hospital, University of Turku, 20520 Turku, Finland;
| | - Tomi T. Laitinen
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, 20520 Turku, Finland; (L.A.M.); (T.T.L.); (J.N.); (S.P.R.); (O.T.R.); (K.P.)
- Centre for Population Health Research, Turku University Hospital, University of Turku, 20520 Turku, Finland;
- Paavo Nurmi Centre, Sports & Exercise Medicine Unit, Department of Physical Activity and Health, University of Turku, 20520 Turku, Finland
| | - Joel Nuotio
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, 20520 Turku, Finland; (L.A.M.); (T.T.L.); (J.N.); (S.P.R.); (O.T.R.); (K.P.)
- Centre for Population Health Research, Turku University Hospital, University of Turku, 20520 Turku, Finland;
- Heart Center, Turku University Hospital, University of Turku, 20520 Turku, Finland
| | - Harri Niinikoski
- Department of Pediatrics and Adolescent Medicine, Turku University Hospital, University of Turku, 20520 Turku, Finland;
| | - Suvi P. Rovio
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, 20520 Turku, Finland; (L.A.M.); (T.T.L.); (J.N.); (S.P.R.); (O.T.R.); (K.P.)
- Centre for Population Health Research, Turku University Hospital, University of Turku, 20520 Turku, Finland;
| | - Hanna Lagström
- Centre for Population Health Research, Turku University Hospital, University of Turku, 20520 Turku, Finland;
- Department of Public Health, Turku University Hospital, University of Turku, 20520 Turku, Finland
| | - Jorma S. A. Viikari
- Division of Medicine, Department of Medicine, Turku University Hospital, University of Turku, 20520 Turku, Finland; (J.S.A.V.); (T.R.)
| | - Tapani Rönnemaa
- Division of Medicine, Department of Medicine, Turku University Hospital, University of Turku, 20520 Turku, Finland; (J.S.A.V.); (T.R.)
| | - Antti Jula
- Department of Chronic Disease Prevention, Institute for Health and Welfare, 20750 Turku, Finland;
| | - Mika Ala-Korpela
- Computational Medicine, Faculty of Medicine, University of Oulu & Biocenter Oulu, 90014 Oulu, Finland;
- NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, 70210 Kuopio, Finland
| | - Olli T. Raitakari
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, 20520 Turku, Finland; (L.A.M.); (T.T.L.); (J.N.); (S.P.R.); (O.T.R.); (K.P.)
- Centre for Population Health Research, Turku University Hospital, University of Turku, 20520 Turku, Finland;
- Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, University of Turku, 20520 Turku, Finland
| | - Katja Pahkala
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, 20520 Turku, Finland; (L.A.M.); (T.T.L.); (J.N.); (S.P.R.); (O.T.R.); (K.P.)
- Centre for Population Health Research, Turku University Hospital, University of Turku, 20520 Turku, Finland;
- Paavo Nurmi Centre, Sports & Exercise Medicine Unit, Department of Physical Activity and Health, University of Turku, 20520 Turku, Finland
| |
Collapse
|
147
|
Mikkola TM, Salonen MK, Kajantie E, Kautiainen H, Eriksson JG. Associations of Fat and Lean Body Mass with Circulating Amino Acids in Older Men and Women. J Gerontol A Biol Sci Med Sci 2021; 75:885-891. [PMID: 31095700 DOI: 10.1093/gerona/glz126] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Indexed: 12/16/2022] Open
Abstract
Circulating amino acids are potential markers of body composition. Previous studies are mainly limited to middle age and focus on either fat or lean mass, thereby ignoring overall body composition. We investigated the associations of fat and lean body mass with circulating amino acids in older men and women. We studied 594 women and 476 men from the Helsinki Birth Cohort Study (age 62-74 years). Bioelectrical impedance analysis was used to indicate two main body compartments by fat (fat mass/height2) and lean mass indices (lean mass/height2), dichotomized based on sex-specific medians. Eight serum amino acids were quantified using nuclear magnetic resonance spectroscopy. General linear models were adjusted for age, smoking, and fasting glucose. Higher lean mass index (LMI) was associated with higher concentrations of branched-chain amino acids in both sexes (p ≤ .001). In men, LMI was also positively associated with tyrosine (p = .006) and inversely with glycine (p < .001). Higher fat mass index was associated with higher concentrations of all branched-chain amino acids, aromatic amino acids (phenylalanine and tyrosine), and alanine in both sexes (p ≤ .008). Associations between body composition and amino acids are largely similar in older men and women. The associations are largely similar to those previously observed in younger adults.
Collapse
Affiliation(s)
| | - Minna K Salonen
- Folkhälsan Research Center, Helsinki, Finland.,Public Health Promotion Unit, National Institute for Health and Welfare, Helsinki, Finland
| | - Eero Kajantie
- Public Health Promotion Unit, National Institute for Health and Welfare, Helsinki, Finland.,PEDEGO Research Unit, MRC Oulu, Oulu University Hospital and University of Oulu, Finland.,Department of Clinical and Molecular Medicine, Norwegian University for Science and Technology, Trondheim, Norway.,Children's Hospital, Helsinki University Hospital and University of Helsinki, Finland
| | - Hannu Kautiainen
- Folkhälsan Research Center, Helsinki, Finland.,Primary Health Care Unit, Kuopio University Hospital, Finland
| | - Johan G Eriksson
- Folkhälsan Research Center, Helsinki, Finland.,Public Health Promotion Unit, National Institute for Health and Welfare, Helsinki, Finland.,Department of General Practice and Primary Health Care, University of Helsinki and Helsinki University Hospital, Finland.,Singapore Institute for Clinical Sciences, Agency for Science, Technology, and Research.,Department of Obstetrics & Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore
| |
Collapse
|
148
|
Ye Z, Zhang C, Zhao Y. Potential effects of adropin on systemic metabolic and hormonal abnormalities in polycystic ovary syndrome. Reprod Biomed Online 2021; 42:1007-1014. [PMID: 33612434 DOI: 10.1016/j.rbmo.2021.01.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 01/28/2021] [Accepted: 01/28/2021] [Indexed: 12/25/2022]
Abstract
RESEARCH QUESTION The study aimed to explore the effects of adropin on hormone concentrations and glucolipid and amino acid metabolism in patients with polycystic ovary syndrome (PCOS). DESIGN A total of 220 PCOS patients and 67 controls were recruited and basal state plasma was collected for adropin measurement. Follicular fluids from 50 PCOS patients and 30 controls were also collected for adropin analysis. RESULTS Plasma and follicular fluid adropin concentrations were lower in PCOS patients than controls (P < 0.001; P = 0.017). Adropin concentrations were negatively correlated with body mass index (BMI), free androgen index, Homeostatic Model Assessment of Insulin Resistance (HOMA-IR), androstenedione and triglycerides, and positively correlated with sex hormone-binding globulin (SHBG) and high-density lipoprotein (HDL) (all P < 0.05). PCOS patients who were either obese or glucose intolerant had lower adropin concentrations (P < 0.001). Follicular fluid adropin concentrations were negatively correlated with HOMA-IR, isoleucine and valine in the PCOS group (all P < 0.05). After adjusting for age, BMI, HOMA-IR and HDL, SHBG still significantly impacted the concentration of plasma adropin (P = 0.022). After adjusting for age and BMI, the odds of subjects with reduced adropin concentrations having PCOS still remained significant (P = 0.045). The association between plasma adropin and PCOS disappeared following SHBG adjustment. CONCLUSIONS The concentrations of adropin in plasma and follicular fluid were lower in PCOS patients, which might affect the development of insulin resistance. The association between adropin concentrations and hyperandrogenaemia in PCOS might be mediated through SHBG. The decreased adropin concentrations are associated with abnormal branched-chain amino acid metabolism in PCOS.
Collapse
Affiliation(s)
- Zhenhong Ye
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital Beijing, China; National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital) Beijing, China; Key Laboratory of Assisted Reproduction, Ministry of Education (Peking University) Beijing, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology Beijing, China
| | - Chunmei Zhang
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital Beijing, China; National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital) Beijing, China; Key Laboratory of Assisted Reproduction, Ministry of Education (Peking University) Beijing, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology Beijing, China
| | - Yue Zhao
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital Beijing, China; National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital) Beijing, China; Key Laboratory of Assisted Reproduction, Ministry of Education (Peking University) Beijing, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology Beijing, China; Research Units of Comprehensive Diagnosis and Treatment of Oocyte Maturation Arrest, Chinese Academy of Medical Sciences Beijing, China.
| |
Collapse
|
149
|
Abstract
Sensing and responding to changes in nutrient levels, including those of glucose, lipids, and amino acids, by the body is necessary for survival. Accordingly, perturbations in nutrient sensing are tightly linked with human pathologies, particularly metabolic diseases such as obesity, type 2 diabetes mellitus, and other complications of metabolic syndromes. The conventional view is that amino acids are fundamental elements for protein and peptide synthesis, while recent studies have revealed that amino acids are also important bioactive molecules that play key roles in signaling pathways and metabolic regulation. Different pathways that sense intracellular and extracellular levels of amino acids are integrated and coordinated at the organismal level, and, together, these pathways maintain whole metabolic homeostasis. In this review, we discuss the studies describing how important sensing signals respond to amino acid availability and how these sensing mechanisms modulate metabolic processes, including energy, glucose, and lipid metabolism. We further discuss whether dysregulation of amino acid sensing signals can be targeted to promote metabolic disorders, and discuss how to translate these mechanisms to treat human diseases. This review will help to enhance our overall understanding of the correlation between amino acid sensing and metabolic homeostasis, which have important implications for human health.
Collapse
Affiliation(s)
- Xiaoming Hu
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Feifan Guo
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
150
|
Zhang X, Xu D, Chen M, Wang Y, He L, Wang L, Wu J, Yin J. Impacts of Selected Dietary Nutrient Intakes on Skeletal Muscle Insulin Sensitivity and Applications to Early Prevention of Type 2 Diabetes. Adv Nutr 2021; 12:1305-1316. [PMID: 33418570 PMCID: PMC8321846 DOI: 10.1093/advances/nmaa161] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 08/11/2020] [Accepted: 11/13/2020] [Indexed: 11/14/2022] Open
Abstract
As the largest tissue in the body, skeletal muscle not only plays key roles in movement and glucose uptake and utilization but also mediates insulin sensitivity in the body by myokines. Insulin resistance in the skeletal muscle is a major feature of type 2 diabetes (T2D). A weakened response to insulin could lead to muscle mass loss and dysfunction. Increasing evidence in skeletal muscle cells, rodents, nonhuman primates, and humans has shown that restriction of caloric or protein intake positively mediates insulin sensitivity. Restriction of essential or nonessential amino acids was reported to facilitate glucose utilization and regulate protein turnover in skeletal muscle under certain conditions. Furthermore, some minerals, such as zinc, chromium, vitamins, and some natural phytochemicals such as curcumin, resveratrol, berberine, astragalus polysaccharide, emodin, and genistein, have been shown recently to protect skeletal muscle cells, mice, or humans with or without diabetes from insulin resistance. In this review, we discuss the roles of nutritional interventions in the regulation of skeletal muscle insulin sensitivity. A comprehensive understanding of the nutritional regulation of insulin signaling would contribute to the development of tools and treatment programs for improving skeletal muscle health and for preventing T2D.
Collapse
Affiliation(s)
- Xin Zhang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China,State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Doudou Xu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Meixia Chen
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Yubo Wang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Linjuan He
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Lu Wang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Jiangwei Wu
- College of Animal Science and Technology, Northwest Agriculture and Forestry University, Yangling, China
| | | |
Collapse
|