1
|
Filippas-Ntekouan S, Dimou A, Dafopoulos P, Kostara C, Bairaktari E, Chasapi S, Spyroulias G, Koufakis T, Koutsovasilis A, Tsimihodimos V. Effect of dapagliflozin on the serum metabolome in patients with type 2 diabetes mellitus. J Diabetes Metab Disord 2025; 24:4. [PMID: 39697865 PMCID: PMC11649604 DOI: 10.1007/s40200-024-01508-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 12/01/2024] [Indexed: 12/20/2024]
Abstract
Objectives SGLT-2 inhibitors have been shown to exert cardio- and renoprotective actions. We aimed to investigate the underlying mechanisms using 1H-NMR based metabolomics in patients with type-2 diabetes mellitus who received dapagliflozin. Methods 50 patients with type 2 diabetes mellitus, inadequately controlled on metformin monotherapy (HbA1c > 7%) received dapagliflozin for 3 months and 30 matched patients received insulin degludec for 3 months. Clinical and laboratory values, as well as 1H-NMR based metabolomics were assessed before treatment and after completion of 3 months of treatment. Results Dapagliflozin reduced weight, body mass index, systolic and diastolic blood pressure significantly. Using 1H-NMR based metabolomics, the dapagliflozin group showed a good separation with a degree of overlap before and after treatment initiation. Regarding targeted metabolomics, dapagliflozin increased serum ketone, citrate and tryptophan levels compared with insulin. On the other hand, serum taurine, threonine and mannose levels were significantly decreased following dapagliflozin administration. Conclusions Dapagliflozin led to a small, but significant change in serum metabolome. The observed changes may indicate improvement in energy metabolism, reduction in inflammatory activity and decreased insulin resistance which may provide further evidence of the agent's observed cardiac and renal protection. The study was registered with ClinicalTrials.gov (identifier: NCT02798757). Supplementary Information The online version contains supplementary material available at 10.1007/s40200-024-01508-1.
Collapse
Affiliation(s)
| | - Aikaterini Dimou
- Laboratory of Clinical Chemistry, University of Ioannina, Ioannina, Greece
| | | | - Christina Kostara
- Laboratory of Clinical Chemistry, University of Ioannina, Ioannina, Greece
| | - Eleni Bairaktari
- Laboratory of Clinical Chemistry, University of Ioannina, Ioannina, Greece
| | | | | | - Theoharis Koufakis
- 2nd Propedeutic Department of Internal Medicine, Aristotle University of Thessaloniki, Hippokration General Hospital, Thessaloniki, 546 42 Greece
| | | | - Vasileios Tsimihodimos
- Department of Internal Medicine, University of Ioannina, Stavrou Niarchou Avenue, Ioannina, 45500 Greece
| |
Collapse
|
2
|
Yu Y, Yang X, Deng J, Yin Y, Wu Y, Yu R. Association of the gut microbiome with diabetic nephropathy and the mediated effect of metabolites: friend or enemy? Int Urol Nephrol 2025:10.1007/s11255-025-04519-w. [PMID: 40257664 DOI: 10.1007/s11255-025-04519-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Accepted: 04/09/2025] [Indexed: 04/22/2025]
Abstract
OBJECTIVE The effects of gut microbiome and its metabolites on diabetic nephropathy (DN) have been inadequately elucidated. The aim of this study is to assess the causal effect of gut microbiome on DN and the mediated effect of metabolites by a two-step Mendelian randomization (MR). METHODS Datasets of gut microbiome, metabolites, and DN were acquired in genome-wide association studies and screened for single nucleotide polymorphisms according to the underlying assumptions of MR. Subsequently, inverse variance weighted was used as the primary method for MR analysis to assess the causal effect of gut microbiome on DN and the mediated effect of metabolites. Finally, MR-Egger intercept, Cochran's Q test, and leave-one-out sensitivity analysis were used to assess the horizontal pleiotropy, heterogeneity, and robustness of the results, respectively. RESULTS The MR analysis demonstrated that Parabacteroides merdae increased the genetic susceptibility to DN by reducing acetylcarnitine (C2) to propionylcarnitine (C3) ratio (mediated proportion 8.95%, mediated effect 0.024) and alpha-ketobutyrate to 3-methyl-2-oxovalerate ratio (mediated proportion 19.90%, mediated effect 0.053). MR Egger showed that these results lack horizontal pleiotropy (p ≥ 0.05). Cochran's Q and sensitivity analysis suggested these results had no heterogeneity (p ≥ 0.05) and were robust. CONCLUSION Our findings revealed the pathway by which Parabacteroides merdae increased the genetic susceptibility to DN by regulating acetylcarnitine (C2) to propionylcarnitine (C3) ratio and alpha-ketobutyrate to 3-methyl-2-oxovalerate ratio. It provides new genetic insights for understanding the pathogenesis of DN and related drug research.
Collapse
Affiliation(s)
- Yunfeng Yu
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Xinyu Yang
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Juan Deng
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Yuman Yin
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Yongjun Wu
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan, China.
| | - Rong Yu
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China.
- The First Hospital, Hunan University of Chinese Medicine, Changsha, Hunan, China.
| |
Collapse
|
3
|
Song J, Wang C, Zhao T, Zhang Y, Xing J, Zhao X, Zhang Y, Zhang Z. Multi-omics approaches for biomarker discovery and precision diagnosis of prediabetes. Front Endocrinol (Lausanne) 2025; 16:1520436. [PMID: 40162315 PMCID: PMC11949806 DOI: 10.3389/fendo.2025.1520436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 02/24/2025] [Indexed: 04/02/2025] Open
Abstract
Recent advancements in multi-omics technologies have provided unprecedented opportunities to identify biomarkers associated with prediabetes, offering novel insights into its diagnosis and management. This review synthesizes the latest findings on prediabetes from multiple omics domains, including genomics, epigenomics, transcriptomics, proteomics, metabolomics, microbiomics, and radiomics. We explore how these technologies elucidate the molecular and cellular mechanisms underlying prediabetes and analyze potential biomarkers with predictive value in disease progression. Integrating multi-omics data helps address the limitations of traditional diagnostic methods, enabling early detection, personalized interventions, and improved patient outcomes. However, challenges such as data integration, standardization, and clinical validation and translation remain to be resolved. Future research leveraging artificial intelligence and machine learning is expected to further enhance the predictive power of multi-omics technologies, contributing to the precision diagnosis and tailored management of prediabetes.
Collapse
Affiliation(s)
- Jielin Song
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- TCM Institute of Sore and Ulcer, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Institute of Traditional Chinese Medicine Surgery, Tianjin, China
| | - Chuanfu Wang
- Department of Encephalopathy, Liangping District Hospital of Traditional Chinese Medicine, Chongqing, China
| | - Tong Zhao
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- TCM Institute of Sore and Ulcer, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Institute of Traditional Chinese Medicine Surgery, Tianjin, China
| | - Yu Zhang
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- TCM Institute of Sore and Ulcer, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Institute of Traditional Chinese Medicine Surgery, Tianjin, China
| | - Jixiang Xing
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- TCM Institute of Sore and Ulcer, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Institute of Traditional Chinese Medicine Surgery, Tianjin, China
| | - Xuelian Zhao
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- TCM Institute of Sore and Ulcer, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Institute of Traditional Chinese Medicine Surgery, Tianjin, China
| | - Yunsha Zhang
- TCM Institute of Sore and Ulcer, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Institute of Traditional Chinese Medicine Surgery, Tianjin, China
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Zhaohui Zhang
- TCM Institute of Sore and Ulcer, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Institute of Traditional Chinese Medicine Surgery, Tianjin, China
- Department of Traditional Chinese Medicine Surgery, The Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
4
|
Huang Y, Liu W, Song G, Wu S, Li X, Shen G, Feng J. Metabolomic analyses of multiple biologic matrices reveal metabolic heterogeneity in diabetic complications. Acta Diabetol 2025:10.1007/s00592-025-02481-8. [PMID: 40080196 DOI: 10.1007/s00592-025-02481-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Accepted: 02/27/2025] [Indexed: 03/15/2025]
Abstract
AIMS Type 2 diabetes mellitus (T2DM) arises from a complex interplay of genetic and environmental factors. Patients with T2DM are susceptible to hyperglycemia-related complications that can impair organ function, underscoring the need to explore the metabolic profiles of affected organs. METHODS In this study, a comprehensive metabolomic analysis was conducted on the serum, kidney, and heart tissues from a rat model of diabetic complications (DC). Pattern recognition and multivariate statistical analyses were applied to identify the potential biomarkers of DC, and metabolic network analysis served to understand the specific metabolic pathways associated with DC. RESULTS Fourteen significantly altered metabolites were identified in serum, 20 in the kidney, and 14 in the heart. The corresponding metabolic pathways included mineral absorption, mTOR signaling pathway, taurine and hypotaurine metabolism, glycine, serine and threonine metabolism, ABC transporters, glucagon signaling pathway, protein degradation and uptake, galactose metabolism, purine metabolism, nicotinic acid and nicotinamide metabolism, and glycolysis and gluconeogenesis. Differential metabolite network analysis revealed instinct metabolic patterns among the serum, kidney, and heart. Notably, the serum's metabolic correlation patterns were found to be somewhat similar to those observed in the kidney, whereas the heart exhibited less pronounced metabolite correlations compared to the other two biological matrices. CONCLUSIONS These findings provide insights into the mechanism underlying the development of diabetic complications. The integration of metabolomics and biological network analyses into diabetes research can potentially revolutionize the field by revealing novel biomarkers for early detection and personalized treatment of diabetes and its associated complications.
Collapse
Affiliation(s)
- Yao Huang
- Department of Electronic Science, Fujian Provincial Key Laboratory of Plasma and Magnetic Resonance, Xiamen University, 422 Siming South Road, Siming District, Xiamen, 361005, Fujian, China
| | - Wuping Liu
- Department of Electronic Science, Fujian Provincial Key Laboratory of Plasma and Magnetic Resonance, Xiamen University, 422 Siming South Road, Siming District, Xiamen, 361005, Fujian, China
| | - Ge Song
- Department of Electronic Science, Fujian Provincial Key Laboratory of Plasma and Magnetic Resonance, Xiamen University, 422 Siming South Road, Siming District, Xiamen, 361005, Fujian, China
| | - Sheng Wu
- Department of Electronic Science, Fujian Provincial Key Laboratory of Plasma and Magnetic Resonance, Xiamen University, 422 Siming South Road, Siming District, Xiamen, 361005, Fujian, China
| | - Xuejun Li
- The Xiamen Diabetes Institute and Department of Endocrinology and Diabetes, the First Affiliated Hospital of Xiamen University, Xiamen, 361003, China
| | - Guiping Shen
- Department of Electronic Science, Fujian Provincial Key Laboratory of Plasma and Magnetic Resonance, Xiamen University, 422 Siming South Road, Siming District, Xiamen, 361005, Fujian, China
| | - Jianghua Feng
- Department of Electronic Science, Fujian Provincial Key Laboratory of Plasma and Magnetic Resonance, Xiamen University, 422 Siming South Road, Siming District, Xiamen, 361005, Fujian, China.
| |
Collapse
|
5
|
Abushamat LA, Yu B, Hoogeveen RC, Sun C, Cheng C, Hartig SM, Herman MA, Balasubramanyam A, Reusch JE, Selvin E, Ndumele CE, Nambi V, Ballantyne CM. Erythritol, Erythronate, and Cardiovascular Outcomes in Older Adults in the ARIC Study. JACC. ADVANCES 2025; 4:101605. [PMID: 39983608 PMCID: PMC11889355 DOI: 10.1016/j.jacadv.2025.101605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 01/08/2025] [Accepted: 01/10/2025] [Indexed: 02/23/2025]
Abstract
BACKGROUND Circulating erythritol, an endogenously produced metabolite and an artificial sweetener, is associated with cardiovascular outcomes. OBJECTIVES The authors assessed associations of erythritol and its downstream metabolite, erythronate, with cardiovascular risk factors and events in older adults in the ARIC (Atherosclerosis Risk In Communities) study (visit 5, 2011-2013). METHODS We included 4,006 participants without prevalent cardiovascular disease and with metabolomic profiling. Erythritol and erythronate were measured by mass spectrometry. We analyzed associations of log-transformed erythritol and erythronate with cardiovascular risk factors and events using Cox proportional hazard models. RESULTS Participants in the highest tertiles of erythritol or erythronate were older, more likely to have diabetes, hypertension, hyperlipidemia, or microalbuminuria, and had higher body mass index and cardiac biomarkers and lower estimated glomerular filtration rate (P < 0.001). Over median follow-up of 8.41 (7.62, 8.93) years, higher erythritol and erythronate concentrations were significantly associated with heart failure (HF) hospitalization, HF with preserved ejection fraction, cardiovascular death, and total mortality after adjustment for demographics and traditional cardiovascular risk factors. Erythronate was additionally significantly associated with coronary heart disease (HR: 1.30 [95% CI: 1.04-1.61], P = 0.02), stroke (1.40 [95% CI: 1.08-1.83], P = 0.012), and HF with reduced ejection fraction (1.38 [95% CI: 1.09-1.74], P = 0.007). Diabetes status did not modify any of these associations (P for interaction >0.20). CONCLUSIONS Circulating erythritol and erythronate levels are markers of cardiometabolic health and cardiovascular outcomes in an older adult population. In particular, erythronate is associated with all cardiovascular outcomes assessed. Future studies should assess the role of erythronate and its related pathways in cardiovascular disease.
Collapse
Affiliation(s)
- Layla A Abushamat
- Section of Cardiovascular Research, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA.
| | - Bing Yu
- Department of Epidemiology, School of Public Health, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Ron C Hoogeveen
- Section of Cardiovascular Research, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Caroline Sun
- Section of Cardiovascular Research, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Chao Cheng
- Section of Epidemiology and Population Science, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Sean M Hartig
- Section of Endocrinology, Diabetes and Metabolism, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Mark A Herman
- Section of Endocrinology, Diabetes and Metabolism, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Ashok Balasubramanyam
- Section of Endocrinology, Diabetes and Metabolism, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Jane Eb Reusch
- Division of Endocrinology, Metabolism, and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA; Rocky Mountain Regional VAMC, Aurora, Colorado, USA
| | - Elizabeth Selvin
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Chiadi E Ndumele
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA; Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Vijay Nambi
- Section of Cardiovascular Research, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA; Section of Cardiology, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Christie M Ballantyne
- Section of Cardiovascular Research, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA; Section of Cardiology, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
6
|
Chen L, Chen B, Dai Y, Sun Q, Wu J, Zheng D, Vgontzas AN, Tang X, Li Y. The association of objective daytime sleepiness with impaired glucose metabolism in patients with obstructive sleep apnea: a multi-omics study. Sleep 2025; 48:zsae240. [PMID: 39549285 DOI: 10.1093/sleep/zsae240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 09/20/2024] [Indexed: 11/18/2024] Open
Abstract
STUDY OBJECTIVES To examine the joint effect of obstructive sleep apnea (OSA) and objective excessive daytime sleepiness (EDS) on glucose metabolism and the underlying mechanisms. METHODS We included 127 patients with OSA. The multiple sleep latency test (MSLT) and Epworth sleepiness scale (ESS) were used to assess objective and subjective EDS, respectively. Disordered glucose metabolism was defined as either a physician diagnosis or having fasting blood glucose levels ≥5.6 mmol/L. Values of fasting insulin and homeostasis model assessment of insulin resistance (HOMA-IR) higher than the median values of our sample were defined as high fasting insulin and insulin resistance. Serum metabolomics and fecal microbiota were used to explore underlying mechanisms. RESULTS Lower MSLT values were associated with higher levels of fasting blood glucose, fasting insulin, and HOMA-IR. Furthermore, objective EDS was associated with increased odds of disordered glucose metabolism, elevated fasting insulin, and insulin resistance. Dysregulation of serum valine degradation and dysbiosis of fecal Bacteroides thetaiotaomicron were associated with impaired glucose metabolism in OSA with objective EDS. No association between subjective EDS and impaired glucose metabolism was observed. CONCLUSIONS OSA with objective, but not subjective, EDS is associated with an increased risk of disordered glucose metabolism and insulin resistance. Dysregulation of valine degradation and dysbiosis of B. thetaiotaomicron appear to link objective EDS and disordered glucose metabolism in OSA.
Collapse
Affiliation(s)
- Le Chen
- Department of Sleep Medicine, Shantou University Mental Health Center, Shantou University Medical College, Shantou, China
- Sleep Medicine Center, Shantou University Medical College, Shantou, China
- Joint Lab of Biological Psychiatry Shantou University-University of Manitoba, Shantou University Medical College, Shantou, China
| | - Baixin Chen
- Department of Sleep Medicine, Shantou University Mental Health Center, Shantou University Medical College, Shantou, China
- Sleep Medicine Center, Shantou University Medical College, Shantou, China
- Joint Lab of Biological Psychiatry Shantou University-University of Manitoba, Shantou University Medical College, Shantou, China
| | - Yanyuan Dai
- Department of Sleep Medicine, Shantou University Mental Health Center, Shantou University Medical College, Shantou, China
- Sleep Medicine Center, Shantou University Medical College, Shantou, China
- Joint Lab of Biological Psychiatry Shantou University-University of Manitoba, Shantou University Medical College, Shantou, China
| | - Qimeng Sun
- Department of Sleep Medicine, Shantou University Mental Health Center, Shantou University Medical College, Shantou, China
| | - Jun Wu
- Department of Sleep Medicine, Shantou University Mental Health Center, Shantou University Medical College, Shantou, China
- Sleep Medicine Center, Shantou University Medical College, Shantou, China
- Joint Lab of Biological Psychiatry Shantou University-University of Manitoba, Shantou University Medical College, Shantou, China
| | - Dandan Zheng
- Department of Sleep Medicine, Shantou University Mental Health Center, Shantou University Medical College, Shantou, China
- Sleep Medicine Center, Shantou University Medical College, Shantou, China
- Joint Lab of Biological Psychiatry Shantou University-University of Manitoba, Shantou University Medical College, Shantou, China
| | - Alexandros N Vgontzas
- Department of Psychiatry and Behavioral Health, Sleep Research and Treatment Center, Pennsylvania State University, College of Medicine, Hershey, PA, USA
| | - Xiangdong Tang
- Sleep Medicine Center, Mental Health Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yun Li
- Department of Sleep Medicine, Shantou University Mental Health Center, Shantou University Medical College, Shantou, China
- Sleep Medicine Center, Shantou University Medical College, Shantou, China
- Joint Lab of Biological Psychiatry Shantou University-University of Manitoba, Shantou University Medical College, Shantou, China
| |
Collapse
|
7
|
Qi Y, Li X, Liu Y, Wang M, Wu Y, Su Q, Qin L, Ma J. Elevated fasting serum xylitol levels are associated with a lower risk of incident type 2 diabetes among individuals with prediabetes in the Chinese population. Nutr Diabetes 2025; 15:3. [PMID: 39910054 PMCID: PMC11799369 DOI: 10.1038/s41387-025-00357-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 12/18/2024] [Accepted: 01/22/2025] [Indexed: 02/07/2025] Open
Abstract
BACKGROUND It is important to detect the predictors of prediabetes progressing to diabetes. Although polyols affect glycometabolism, little is known about the association between fasting serum polyol levels of participants with habitual diet and the risk of prediabetes progressing to type 2 diabetes. METHODS In this nested case-control study, 180 participants who developed from prediabetes to type 2 diabetes (progressors), and 180 matched controls (non-progressors) with prediabetes during a 3.5-year follow-up were enrolled. The baseline levels of serum polyols in the fasting state were quantified using time-of-flight mass spectrometry. Multivariate conditional logistic regression was performed to assess the effects of the differential polyol levels on the risk of incident diabetes from prediabetes. RESULTS The baseline fasting xylitol levels, but not sorbitol or erythritol levels, were higher in non-progressors than in progressors (P < 0.001). Non-progressors, in comparison with progressors, had significantly higher proportions within the third tertile of xylitol levels (71/180 non-progressors [39.4%] vs. 49/180 progressors [27.2%]). After adjusting for potential confounders, the odds ratio of risk for incident diabetes in the highest tertile of xylitol levels was 0.338 (95% confidence interval 0.182-0.628), when compared with those in the lowest tertile. In addition, the association between xylitol levels and incident diabetes was persistent in those with fasting hyperglycemia and both fasting and 2h-post-glucose-load hyperglycemia, but not in the isolated 2h-post-glucose-load hyperglycemia. CONCLUSIONS Elevated baseline fasting serum xylitol levels are associated with a lower risk of prediabetes progressing to diabetes. This association was particularly evident in those with fasting hyperglycemia. These findings suggest that fasting serum xylitol levels may serve as an important predictor and protective factor against the development of diabetes.
Collapse
Affiliation(s)
- Yicheng Qi
- Department of Endocrinology and Metabolism, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoyong Li
- Department of Endocrinology and Metabolism, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Liu
- Department of Endocrinology and Metabolism, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Meng Wang
- Department of Endocrinology and Metabolism, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yiming Wu
- Department of Endocrinology, Chongming Hospital Affiliated to Shanghai University of Health & Medicine Sciences (Chongming Branch of Xinhua Hospital), Shanghai, China
| | - Qing Su
- Department of Endocrinology and Metabolism, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Li Qin
- Department of Endocrinology and Metabolism, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Department of Endocrinology, Chongming Hospital Affiliated to Shanghai University of Health & Medicine Sciences (Chongming Branch of Xinhua Hospital), Shanghai, China.
| | - Jing Ma
- Department of Endocrinology and Metabolism, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
8
|
McCann JR, Yang C, Bihlmeyer N, Tang R, Truong T, An J, Jawahar J, Ilkayeva O, Muehlbauer M, Hu ZZ, Dressman H, Poppe L, Granek J, David LA, Shi P, Balikcioglu PG, Shah S, Armstrong SC, Newgard CB, Seed PC, Rawls JF. Branched chain amino acid metabolism and microbiome in adolescents with obesity during weight loss therapy. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.02.03.25321363. [PMID: 39974080 PMCID: PMC11838640 DOI: 10.1101/2025.02.03.25321363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
BACKGROUND Towards improving outcomes for adolescents with obesity, we aimed to define metabolic and microbiome phenotypes at baseline and post-weight loss intervention. METHODS The Pediatric Obesity Microbiome and Metabolism Study enrolled 220 adolescents aged 10-18 with severe obesity (OB) and 67 healthy weight controls (HWC). Blood, stool, and clinical measures were collected at baseline and after a 6-month intervention for the OB group. Serum metabolomic and fecal microbiome data were analyzed for associations with BMI, insulin resistance, and inflammation. Fecal microbiome transplants were performed on germ-free mice using samples from both groups to assess weight gain and metabolomic changes. RESULTS Adolescents with OB exhibited elevated serum branched-chain amino acids (BCAA) but reduced ketoacid metabolites (BCKA) compared to HWC. This pattern was sex- and age-dependent, unlike adults with OB, who showed elevated levels of both. The fecal microbiomes of adolescents with OB and HWC had similar diversity but differed in membership and functional potential. FMT from OB and HWC donors had similar effects on mouse body weight, with specific taxa linked to weight gain in FMT recipients. Longitudinal analysis identified metabolic and microbial features correlated with changes in health measures during the intervention. CONCLUSION Adolescents with OB have unique metabolomic adaptations and microbiome signatures compared to their HWC counterparts and adults with OB. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT03139877 (Observational Study) and NCT02959034 (Repository). FUNDING SOURCES American Heart Association Grants: 17SFRN33670990, 20PRE35180195National Institute of Diabetes and Digestive and Kidney Diseases Grant: R24-DK110492.
Collapse
|
9
|
Wei X, Wei S, Chen M, Tan Y, Yang Z, Feng W, Yang G, Han Z, Luo X. Subcutaneous adipose tissue compensates for the perturbations in circulating one-carbon metabolism in women with gestational diabetes. Acta Diabetol 2025:10.1007/s00592-025-02452-z. [PMID: 39899132 DOI: 10.1007/s00592-025-02452-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 01/05/2025] [Indexed: 02/04/2025]
Abstract
The prevalence of gestational diabetes mellitus (GDM) is rising and poses important health risks for the mother, developing fetus and offspring, even when maternal glycemic control is well managed. This study aimed to identify the differently expressed metabolites (DEMs) in maternal plasma between GDM pregnancies with good glycemic control and healthy pregnancies, along with the DEMs-related metabolism in adipose tissue. Pregnant women with scheduled caesarean sections were recruited. Venous blood samples were collected on the day prior to delivery for targeted metabolomics analysis focusing on the 200 polar metabolites in central carbon metabolism. Subcutaneous and omental white adipose tissue (sWAT and oWAT) were harvested at delivery. A total of 162 metabolites were quantified, revealing 2 up-regulated (D-glucose 6-phosphate (G6P), succinate) and 8 down-regulated DEMs, which exhibited a fold change of ≥ 1.5 or ≤ 0.67, respectively. Among the down-regulated DEMs, 5 metabolites-pyridoxine, glycine, S-methyl-L-cysteine, methionine, and S-carboxymethyl-L-cysteine-are related to one-carbon metabolism (OCM). In response to perturbation in circulating OCM, boosted methionine cycle, NAD + metabolism, and adipogenesis were observed in sWAT of GDM subjects, with no changes detected in oWAT. None of the 10 DEMs correlates with either blood glucose or insulin, but showed significant correlations with TG, TC, LDL-C and HDL-C. The present study indicates that sWAT compensates for the perturbations in circulating OCM associated with GDM and targeting to the OCM may be an effective strategy to control the long-term metabolic risk of GDM offsprings.
Collapse
Affiliation(s)
- Xiaojing Wei
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 Yanta Western Road, Xi'an, 710061, Shaanxi, China
- Institute of Neuroscience, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education of China, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Shuangyu Wei
- Clinical Laboratory, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Miao Chen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Yutian Tan
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 Yanta Western Road, Xi'an, 710061, Shaanxi, China
- Institute of Neuroscience, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education of China, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Zhao Yang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Weijie Feng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 Yanta Western Road, Xi'an, 710061, Shaanxi, China
- Institute of Neuroscience, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education of China, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Guiying Yang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Zhen Han
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Xiao Luo
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 Yanta Western Road, Xi'an, 710061, Shaanxi, China.
- Institute of Neuroscience, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China.
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education of China, Xi'an Jiaotong University, Xi'an, 710061, China.
| |
Collapse
|
10
|
Zhao Y, Chai X, Peng J, Zhu Y, Dong R, He J, Xia L, Liu S, Chen J, Xu Z, Luo C, Sheng J. Proline exacerbates hepatic gluconeogenesis via paraspeckle-dependent mRNA retention. Nat Metab 2025; 7:367-382. [PMID: 39820557 DOI: 10.1038/s42255-024-01206-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 12/10/2024] [Indexed: 01/19/2025]
Abstract
Type 2 diabetes (T2D) is a global health issue characterized by abnormal blood glucose levels and is often associated with excessive hepatic gluconeogenesis. Increased circulating non-essential amino acids (NEAAs) are consistently observed in individuals with T2D; however, the specific contribution of each amino acid to T2D pathogenesis remains less understood. Here, we report an unexpected role of the NEAA proline in coordinating hepatic glucose metabolism by modulating paraspeckle, a nuclear structure scaffolded by the long non-coding RNA Neat1. Mechanistically, proline diminished paraspeckles in hepatocytes, liberating the retained mRNA species into cytoplasm for translation, including the mRNAs of Ppargc1a and Foxo1, contributing to enhanced gluconeogenesis and hyperglycaemia. We further demonstrated that the proline-paraspeckle-mRNA retention axis existed in diabetic liver samples, and intervening in this axis via paraspeckle restoration substantially alleviated hyperglycaemia in both female and male diabetic mouse models. Collectively, our results not only delineated a previously unappreciated proline-instigated, paraspeckle-dependent mRNA-retention mechanism regulating gluconeogenesis, but also spotlighted proline and paraspeckle as potential targets for managing hyperglycaemia.
Collapse
Affiliation(s)
- Yurong Zhao
- Institute of Environmental Medicine and Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Liangzhu Laboratory, Zhejiang University, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
| | - Xinxin Chai
- Institute of Environmental Medicine and Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Liangzhu Laboratory, Zhejiang University, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
| | - Junxuan Peng
- Institute of Environmental Medicine and Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Liangzhu Laboratory, Zhejiang University, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
| | - Yi Zhu
- Institute of Environmental Medicine and Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Liangzhu Laboratory, Zhejiang University, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
| | - Rong Dong
- NHC Key Laboratory of Pulmonary Immunological Diseases, Guizhou Provincial People's Hospital, Guiyang, China
| | - Junwei He
- College of Life Science, Zhejiang University, Hangzhou, China
| | - Linghao Xia
- College of Life Science, Zhejiang University, Hangzhou, China
| | - Sishuo Liu
- Institute of Environmental Medicine and Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Liangzhu Laboratory, Zhejiang University, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
| | - Jingzhou Chen
- Institute of Environmental Medicine and Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Liangzhu Laboratory, Zhejiang University, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
| | - Zhengping Xu
- Institute of Environmental Medicine and Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Liangzhu Laboratory, Zhejiang University, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
| | - Chi Luo
- Liangzhu Laboratory, Zhejiang University, Hangzhou, China.
| | - Jinghao Sheng
- Institute of Environmental Medicine and Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Liangzhu Laboratory, Zhejiang University, Hangzhou, China.
- Cancer Center, Zhejiang University, Hangzhou, China.
| |
Collapse
|
11
|
Schoumacher M, Lambert V, Campas M, Blaise P, Locht B, Thys M, Duchateau E, Cavalier E, Rakic JM, Noël A, de Tullio P. Opportunities, challenges, and difficulties in NMR-based metabolomics applied to neovascular age-related macular degeneration (nAMD) patient follow-up. Front Mol Biosci 2025; 11:1449226. [PMID: 39935708 PMCID: PMC11811626 DOI: 10.3389/fmolb.2024.1449226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 12/12/2024] [Indexed: 02/13/2025] Open
Abstract
Introduction This study applies NMR-based metabolomics to investigate neovascular age-related macular degeneration (nAMD), addressing challenges in patient management, disease progression evaluation, and treatment response assessment. A two-year follow-up of 29 nAMD patients undergoing treatment provided 231 time points for analysis. Methods Over the two-year period, 11 males and 18 females (aged 61-92 years) were monitored, yielding 231 time points. At each time point, blood samples for NMR metabolomics analysis, clinical measurements (e.g., lactate, glucose levels, HDL/LDL cholesterol, and blood pH), and optical coherence tomography (OCT) images were collected to evaluate the progression of choroidal neovascularization. 1H-NMR metabolomic analysis led to the quantification of over 60 metabolites and of the major lipoprotein fractions. Both multivariate and univariate statistical approaches tailored for longitudinal data were employed to identify biomarkers correlating metabolomic changes with ocular alterations during disease progression. Results and Discussion Despite a rigorous analytical workflow enabling precise quantification of over 60 metabolites and the application of advanced statistical tools for longitudinal data, achieving consistent results across the cohort proved challenging. The dataset's heterogeneity, reflecting real-world clinical practice, complicated the derivation of global conclusions. Personalized analyses on a patient-by-patient basis successfully identified individual correlation models, but a universal model remained elusive. This study highlights the inherent challenges of translating findings from controlled settings into clinical practice, where factors such as visit frequency, treatment variability, and disease heterogeneity limit data uniformity. We emphasize the importance of experimental design in longitudinal studies, particularly when dealing with incomplete and variable datasets. We are therefore confident that, considering both the challenges and difficulties identified in this work and the preliminary results presented here, it is possible to develop predictive and individualized models for monitoring patients with nAMD. Such models could greatly assist clinicians in providing better care for these patients.
Collapse
Affiliation(s)
- M. Schoumacher
- Clinical Metabolomics Group (CliMe), Center for Interdisciplinary Research on Medicines, Université de Liège, Liège, Belgium
- Department of Medical Chemistry, University Hospital of Liège, Liège, Belgium
| | - V. Lambert
- Department of Ophthalmology, University Hospital of Liège, Liège, Belgium
| | - M. Campas
- Clinical Metabolomics Group (CliMe), Center for Interdisciplinary Research on Medicines, Université de Liège, Liège, Belgium
| | - P. Blaise
- Department of Ophthalmology, University Hospital of Liège, Liège, Belgium
| | - B. Locht
- Department of Ophthalmology, University Hospital of Liège, Liège, Belgium
| | - M. Thys
- Department of Ophthalmology, University Hospital of Liège, Liège, Belgium
| | - E. Duchateau
- Department of Ophthalmology, University Hospital of Liège, Liège, Belgium
| | - E. Cavalier
- Clinical Metabolomics Group (CliMe), Center for Interdisciplinary Research on Medicines, Université de Liège, Liège, Belgium
- Department of Medical Chemistry, University Hospital of Liège, Liège, Belgium
| | - J.-M. Rakic
- Department of Ophthalmology, University Hospital of Liège, Liège, Belgium
| | - A. Noël
- Laboratory of Tumor and Development Biology, GIGA, Université de Liège, Liège, Belgium
| | - P. de Tullio
- Clinical Metabolomics Group (CliMe), Center for Interdisciplinary Research on Medicines, Université de Liège, Liège, Belgium
- Department of Medical Chemistry, University Hospital of Liège, Liège, Belgium
| |
Collapse
|
12
|
Khan SR, Ye WW, Van JAD, Singh I, Rabiee Y, Rodricks KL, Zhang X, Nicholson RJ, Razani B, Summers SA, Futerman AH, Gunderson EP, Wheeler MB. Reduced circulating sphingolipids and CERS2 activity are linked to T2D risk and impaired insulin secretion. SCIENCE ADVANCES 2025; 11:eadr1725. [PMID: 39792658 PMCID: PMC11790001 DOI: 10.1126/sciadv.adr1725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 12/09/2024] [Indexed: 01/12/2025]
Abstract
Gestational diabetes mellitus (GDM), a transient form of diabetes that resolves postpartum, is a major risk factor for type 2 diabetes (T2D) in women. While the progression from GDM to T2D is not fully understood, it involves both genetic and environmental components. By integrating clinical, metabolomic, and genome-wide association study (GWAS) data, we identified associations between decreased sphingolipid biosynthesis and future T2D, in part through the rs267738 allele of the CERS2 gene in Hispanic women shortly after a GDM pregnancy. To understand the impact of the CERS2 gene and risk allele on glucose regulation, we examined whole-body Cers2 knockout and rs267738 knock-in mice. Both models exhibited glucose intolerance and impaired insulin secretion in vivo. Islets isolated from these models also demonstrated reduced β cell function, as shown by decreased insulin secretion ex vivo. Overall, reduced circulating sphingolipids may indicate a high risk of GDM-to-T2D progression and reflect deficits in CERS2 activity that negatively affect glucose homeostasis and β cell function.
Collapse
Affiliation(s)
- Saifur R. Khan
- Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- VA Medical Center, Pittsburgh, PA, USA
- Center for Immunometabolism, University of Pittsburgh, Pittsburgh, PA, USA
| | - Wenyue W. Ye
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Julie A. D. Van
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Ishnoor Singh
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Yasmin Rabiee
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | | | - Xiangyu Zhang
- Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- VA Medical Center, Pittsburgh, PA, USA
- Center for Immunometabolism, University of Pittsburgh, Pittsburgh, PA, USA
| | - Rebekah J. Nicholson
- Departments of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA
| | - Babak Razani
- Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- VA Medical Center, Pittsburgh, PA, USA
- Center for Immunometabolism, University of Pittsburgh, Pittsburgh, PA, USA
| | - Scott A. Summers
- Departments of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA
| | - Anthony H. Futerman
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Erica P. Gunderson
- Division of Research, Kaiser Permanente Northern California, Pleasanton, CA, USA
- Department of Health Systems Science, Kaiser Permanente Bernard J. Tyson School of Medicine, Pasadena, CA, USA
| | - Michael B. Wheeler
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
13
|
Zhang Y, Spitzer BW, Zhang Y, Wallace DA, Yu B, Qi Q, Argos M, Avilés-Santa ML, Boerwinkle E, Daviglus ML, Kaplan R, Cai J, Redline S, Sofer T. Untargeted metabolome atlas for sleep-related phenotypes in the Hispanic community health study/study of Latinos. EBioMedicine 2025; 111:105507. [PMID: 39693737 PMCID: PMC11722176 DOI: 10.1016/j.ebiom.2024.105507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 11/25/2024] [Accepted: 12/04/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Sleep is essential to maintaining health and wellbeing of individuals, influencing a variety of outcomes from mental health to cardiometabolic disease. This study aims to assess the relationships between various sleep-related phenotypes and blood metabolites. METHODS Utilising data from the Hispanic Community Health Study/Study of Latinos, we performed association analyses between 40 sleep-related phenotypes, grouped in several domains (sleep disordered breathing (SDB), sleep duration, sleep timing, self-reported insomnia symptoms, excessive daytime sleepiness (EDS), and heart rate during sleep), and 768 metabolites measured via untargeted metabolomics profiling. Network analysis was employed to visualise and interpret the associations between sleep phenotypes and metabolites. FINDINGS The patterns of statistically significant associations between sleep phenotypes and metabolites differed by superpathways, and highlighted subpathways of interest for future studies. For example, primary bile acid metabolism showed the highest cumulative percentage of statistically significant associations across all sleep phenotype domains except for SDB and EDS phenotypes. Several metabolites were associated with multiple sleep phenotypes, from a few domains. Glycochenodeoxycholate, vanillyl mandelate (VMA) and 1-stearoyl-2-oleoyl-GPE (18:0/18:1) were associated with the highest number of sleep phenotypes, while pregnenolone sulfate was associated with all sleep phenotype domains except for sleep duration. N-lactoyl amino acids such as N-lactoyl phenylalanine (lac-Phe), were associated with sleep duration, SDB, sleep timing and heart rate during sleep. INTERPRETATION This atlas of sleep-metabolite associations will facilitate hypothesis generation and further study of the metabolic underpinnings of sleep health. FUNDING R01HL161012, R35HL135818, R01AG80598.
Collapse
Affiliation(s)
- Ying Zhang
- Division of Sleep Medicine and Circadian Disorders, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Brian W Spitzer
- CardioVascular Institute, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Yu Zhang
- CardioVascular Institute, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Danielle A Wallace
- Division of Sleep Medicine and Circadian Disorders, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA; CardioVascular Institute, Beth Israel Deaconess Medical Center, Boston, MA, USA; Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Bing Yu
- Department of Epidemiology, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Qibin Qi
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Maria Argos
- Department of Epidemiology and Biostatistics, School of Public Health, University of Illinois Chicago, Chicago, IL, USA; Department of Environmental Health, School of Public Health, Boston University, Boston, MA, USA
| | - M Larissa Avilés-Santa
- Division of Clinical and Health Services Research, National Institute on Minority Health and Health Disparities, National Institutes of Health, Bethesda, MD, USA
| | - Eric Boerwinkle
- Department of Epidemiology, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Martha L Daviglus
- Institute for Minority Health Research, University of Illinois at Chicago, Chicago, IL, USA
| | - Robert Kaplan
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA; Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Jianwen Cai
- Collaborative Studies Coordinating Center, Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Susan Redline
- Division of Sleep Medicine and Circadian Disorders, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA; Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Tamar Sofer
- Division of Sleep Medicine and Circadian Disorders, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA; CardioVascular Institute, Beth Israel Deaconess Medical Center, Boston, MA, USA; Department of Medicine, Harvard Medical School, Boston, MA, USA; Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA.
| |
Collapse
|
14
|
Shi N, Nepal S, Hoobler R, Menni C, Playdon MC, Spakowicz D, Wells PM, Steves CJ, Clinton SK, Tabung FK. Pro-inflammatory and hyperinsulinaemic dietary patterns are associated with specific gut microbiome profiles: a TwinsUK cohort study. GUT MICROBIOME (CAMBRIDGE, ENGLAND) 2024; 5:e12. [PMID: 39703541 PMCID: PMC11658949 DOI: 10.1017/gmb.2024.14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 06/27/2024] [Accepted: 10/14/2024] [Indexed: 12/21/2024]
Abstract
Metabolic dietary patterns, including the Empirical Dietary Index for Hyperinsulinaemia (EDIH) and Empirical Dietary Inflammatory Pattern (EDIP), are known to impact multiple chronic diseases, but the role of the colonic microbiome in mediating such relationships is poorly understood. Among 1,610 adults with faecal 16S rRNA data in the TwinsUK cohort, we identified the microbiome profiles for EDIH and EDIP (from food frequency questionnaires) cross-sectionally using elastic net regression. We assessed the association of the dietary pattern-related microbiome profile scores with circulating biomarkers in multivariable-adjusted linear regression. In addition, we used PICRUSt2 to predict biological pathways associated with the enriched microbiome profiles, and further screened pathways for associations with the dietary scores in linear regression analyses. Microbiome profile scores developed with 32 (EDIH) and 15 (EDIP) genera were associated with higher insulin and homeostatic model assessment of insulin resistance. Six genera were associated with both dietary scores: Ruminococcaceae_UCG-008, Lachnospiraceae_UCG-008, Defluviitaleaceae_UCG-011 Anaeroplasma, inversely and Negativibacillus, Streptococcus, positively. Further, pathways in fatty acid biosynthesis, sugar acid degradation, and mevalonate metabolism were associated with insulinaemic and inflammatory diets. Dietary patterns that exert metabolic effects on insulin and inflammation may influence chronic disease risk by modulating gut microbial composition and function.
Collapse
Affiliation(s)
- Ni Shi
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Sushma Nepal
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
- Interdisciplinary Ph.D. Program in Nutrition, The Ohio State University, Columbus, OH, USA
| | - Rachel Hoobler
- Department of Nutrition and Integrative Physiology, College of Health, University of Utah, and Huntsman Cancer Institute, Salt Lake City, UT, USA
| | - Cristina Menni
- The Department of Twin Research, Kings College London, London, UK
| | - Mary C. Playdon
- Department of Nutrition and Integrative Physiology, College of Health, University of Utah, and Huntsman Cancer Institute, Salt Lake City, UT, USA
| | - Daniel Spakowicz
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
- Division of Medical Oncology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH, USA
| | | | - Claire J. Steves
- The Department of Twin Research, Kings College London, London, UK
| | - Steven K. Clinton
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
- Interdisciplinary Ph.D. Program in Nutrition, The Ohio State University, Columbus, OH, USA
- Division of Medical Oncology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Fred K. Tabung
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
- Interdisciplinary Ph.D. Program in Nutrition, The Ohio State University, Columbus, OH, USA
- Division of Medical Oncology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH, USA
- Division of Epidemiology, College of Public Health, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
15
|
Amiri-Dashatan N, Etemadi SM, Besharati S, Farahani M, Moghaddam AK. Dysregulation of amino acids balance as potential serum-metabolite biomarkers for diagnosis and prognosis of diabetic retinopathy: a metabolomics study. J Diabetes Metab Disord 2024; 23:2031-2042. [PMID: 39610496 PMCID: PMC11599686 DOI: 10.1007/s40200-024-01462-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Accepted: 06/23/2024] [Indexed: 11/30/2024]
Abstract
Objectives Diabetic retinopathy (DR), an earnest complication of diabetes, is one of the most common causes of blindness worldwide. This study aimed to investigate the altered metabolites in the serum of non-DR (NDR) and DR including non-proliferative diabetic retinopathy (NPDR), and proliferative diabetic retinopathy (PDR) subjects. Methods In this study, the 1HNMR platform was applied to reveal the discriminating serum metabolites in three diabetic groups based on the status of their complications: T2D or NDR (n = 15), NPDR, (n = 15), and PDR (n = 15) groups. Multivariate analyses include principal component analysis (PCA) and Partial Least Structures-Discriminant Analysis (PLS-DA) analysis that were performed using R software. The main metabolic pathways were also revealed by KEGG pathway enrichment analysis. Results The results revealed the significantly different metabolites include 10 metabolites of the NPDR versus PDR group, 24 metabolites of the PDR versus NDR group, and 25 metabolites of the NPDR versus NDR group. The results showed that the significantly altered metabolites in DR compared with NDR serum samples mainly belonged to amino acids. The most important pathways between NPDR/PDR, and NDR/DR groups include ascorbate and aldarate metabolism, galactose metabolism, glutathione metabolism, and tryptophan metabolism, respectively. In addition, some metabolites were detected for the first time. Conclusions We created a metabolomics profile for NDR, PDR and NPDR groups. The impairment in the ascorbate/aldarate, galactose, and especially amino acids metabolism was identified as metabolic dysregulation associated with DR, which may provide new insights into potential pathogenesis pathways for DR. Graphical Abstract
Collapse
Affiliation(s)
- Nasrin Amiri-Dashatan
- Zanjan Metabolic Diseases Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | | | - Shahin Besharati
- Zanjan Metabolic Diseases Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Masoumeh Farahani
- Proteomics Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Arezoo Karimi Moghaddam
- Department of Ophthalmology, School of Medicine, Vali-E-Asr Hospital, Zanjan University of Medical sciences, Zanjan, Iran
| |
Collapse
|
16
|
Li Y, Han S. Metabolomic Applications in Gut Microbiota-Host Interactions in Human Diseases. Gastroenterol Clin North Am 2024; 53:383-397. [PMID: 39068001 DOI: 10.1016/j.gtc.2023.12.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
The human gut microbiota, consisting of trillions of microorganisms, encodes diverse metabolic pathways that impact numerous aspects of host physiology. One key way in which gut bacteria interact with the host is through the production of small metabolites. Several of these microbiota-dependent metabolites, such as short-chain fatty acids, have been shown to modulate host diseases. In this review, we examine how disease-associated metabolic signatures are identified using metabolomic platforms, and where metabolomics is applied in gut microbiota-disease interactions. We further explore how integration of metagenomic and metabolomic data in human studies can facilitate biomarkers discoveries in precision medicine.
Collapse
Affiliation(s)
- Yuxin Li
- Biochemistry Graduate Program, Duke University School of Medicine, Durham, NC 27710, USA
| | - Shuo Han
- Department of Biochemistry, Duke University School of Medicine, Durham, NC 27710, USA; Duke Microbiome Center, Duke University School of Medicine, Durham, NC 27710, USA; Department of Molecular Genetics and Microbiology, Duke University School of Medicine, NC 27710, USA.
| |
Collapse
|
17
|
Pandey S. Metabolomics Characterization of Disease Markers in Diabetes and Its Associated Pathologies. Metab Syndr Relat Disord 2024; 22:499-509. [PMID: 38778629 DOI: 10.1089/met.2024.0038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2024] Open
Abstract
With the change in lifestyle of people, there has been a considerable increase in diabetes, which brings with it certain follow-up pathological conditions, which lead to a substantial medical burden. Identifying biomarkers that aid in screening, diagnosis, and prognosis of diabetes and its associated pathologies would help better patient management and facilitate a personalized treatment approach for prevention and treatment. With the advancement in techniques and technologies, metabolomics has emerged as an omics approach capable of large-scale high throughput data analysis and identifying and quantifying metabolites that provide an insight into the underlying mechanism of the disease and its progression. Diabetes and metabolomics keywords were searched in correspondence with the assigned keywords, including kidney, cardiovascular diseases and critical illness from PubMed and Scopus, from its inception to Dec 2023. The relevant studies from this search were extracted and included in the study. This review is focused on the biomarkers identified in diabetes, diabetic kidney disease, diabetes-related development of CVD, and its role in critical illness.
Collapse
Affiliation(s)
- Swarnima Pandey
- School of Pharmacy, Department of Pharmaceutical Sciences, University of Maryland, Baltimore, Maryland, USA
| |
Collapse
|
18
|
Rakusanova S, Cajka T. Metabolomics and Lipidomics for Studying Metabolic Syndrome: Insights into Cardiovascular Diseases, Type 1 & 2 Diabetes, and Metabolic Dysfunction-Associated Steatotic Liver Disease. Physiol Res 2024; 73:S165-S183. [PMID: 39212142 PMCID: PMC11412346 DOI: 10.33549/physiolres.935443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024] Open
Abstract
Metabolomics and lipidomics have emerged as tools in understanding the connections of metabolic syndrome (MetS) with cardiovascular diseases (CVD), type 1 and type 2 diabetes (T1D, T2D), and metabolic dysfunction-associated steatotic liver disease (MASLD). This review highlights the applications of these omics approaches in large-scale cohort studies, emphasizing their role in biomarker discovery and disease prediction. Integrating metabolomics and lipidomics has significantly advanced our understanding of MetS pathology by identifying unique metabolic signatures associated with disease progression. However, challenges such as standardizing analytical workflows, data interpretation, and biomarker validation remain critical for translating research findings into clinical practice. Future research should focus on optimizing these methodologies to enhance their clinical utility and address the global burden of MetS-related diseases.
Collapse
Affiliation(s)
- S Rakusanova
- Laboratory of Translational Metabolism, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic.
| | | |
Collapse
|
19
|
Beura S, Kundu P, Das AK, Ghosh A. Genome-scale community modelling elucidates the metabolic interaction in Indian type-2 diabetic gut microbiota. Sci Rep 2024; 14:17259. [PMID: 39060274 PMCID: PMC11282233 DOI: 10.1038/s41598-024-63718-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 05/31/2024] [Indexed: 07/28/2024] Open
Abstract
Type-2 diabetes (T2D) is a rapidly growing multifactorial metabolic disorder that induces the onset of various diseases in the human body. The compositional and metabolic shift of the gut microbiota is a crucial factor behind T2D. Hence, gaining insight into the metabolic profile of the gut microbiota is essential for revealing their role in regulating the metabolism of T2D patients. Here, we have focused on the genome-scale community metabolic model reconstruction of crucial T2D-associated gut microbes. The model-based analysis of biochemical flux in T2D and healthy gut conditions showed distinct biochemical signatures and diverse metabolic interactions in the microbial community. The metabolic interactions encompass cross-feeding of short-chain fatty acids, amino acids, and vitamins among individual microbes within the community. In T2D conditions, a reduction in the metabolic flux of acetate, butyrate, vitamin B5, and bicarbonate was observed in the microbial community model, which can impact carbohydrate metabolism. The decline in butyrate levels is correlated with both insulin resistance and diminished glucose metabolism in T2D patients. Compared to the healthy gut, an overall reduction in glucose consumption and SCFA production flux was estimated in the T2D gut environment. Moreover, the decreased consumption profiles of branch chain amino acids (BCAAs) and aromatic amino acids (AAAs) in the T2D gut microbiota can be a distinct biomarker for T2D. Hence, the flux-level analysis of the microbial community model can provide insights into the metabolic reprogramming in diabetic gut microbiomes, which may be helpful in personalized therapeutics and diet design against T2D.
Collapse
Affiliation(s)
- Satyajit Beura
- Department of Bioscience and Biotechnology, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal, 721302, India
| | - Pritam Kundu
- School of Energy Science and Engineering, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal, 721302, India
| | - Amit Kumar Das
- Department of Bioscience and Biotechnology, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal, 721302, India
| | - Amit Ghosh
- School of Energy Science and Engineering, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal, 721302, India.
- P.K. Sinha Centre for Bioenergy and Renewables, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal, 721302, India.
| |
Collapse
|
20
|
Li J, Wang W, Liu F, Qiu L, Ren Y, Li M, Li W, Gao F, Zhang J. Genetically predicted 1091 blood metabolites and 309 metabolite ratios in relation to risk of type 2 diabetes: a Mendelian randomization study. Front Genet 2024; 15:1356696. [PMID: 39050247 PMCID: PMC11266066 DOI: 10.3389/fgene.2024.1356696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Accepted: 06/21/2024] [Indexed: 07/27/2024] Open
Abstract
Background Metabolic dysregulation represents a defining characteristic of Type 2 diabetes (T2DM). Nevertheless, there remains an absence of substantial evidence establishing a direct causal link between circulating blood metabolites and the promotion or prevention of T2DM. In addressing this gap, we employed Mendelian randomization (MR) analysis to investigate the potential causal association between 1,091 blood metabolites, 309 metabolite ratios, and the occurrence of T2DM. Methods Data encompassing single-nucleotide polymorphisms (SNPs) for 1,091 blood metabolites and 309 metabolite ratios were extracted from a Canadian Genome-wide association study (GWAS) involving 8,299 participants. To evaluate the causal link between these metabolites and Type 2 diabetes (T2DM), multiple methods including Inverse Variance Weighted (IVW), Weighted Median, MR Egger, Weighted Mode, and Simple Mode were employed. p-values underwent correction utilizing False Discovery Rates (FDR). Sensitivity analyses incorporated Cochran's Q test, MR-Egger intercept test, MR-PRESSO, Steiger test, leave-one-out analysis, and single SNP analysis. The causal effects were visualized via Circos plot, forest plot, and scatter plot. Furthermore, for noteworthy, an independent T2DM GWAS dataset (GCST006867) was utilized for replication analysis. Metabolic pathway analysis of closely correlated metabolites was conducted using MetaboAnalyst 5.0. Results The IVW analysis method utilized in this study revealed 88 blood metabolites and 37 metabolite ratios demonstrating a significant causal relationship with T2DM (p < 0.05). Notably, strong causal associations with T2DM were observed for specific metabolites: 1-linoleoyl-GPE (18:2) (IVW: OR:0.930, 95% CI: 0.899-0.962, p = 2.16 × 10-5), 1,2-dilinoleoyl-GPE (18:2/18:2) (IVW: OR:0.942, 95% CI: 0.917-0.968, p = 1.64 × 10-5), Mannose (IVW: OR:1.133, 95% CI: 1.072-1.197, p = 1.02 × 10-5), X-21829 (IVW: OR:1.036, 95% CI: 1.036-1.122, p = 9.44 × 10-5), and Phosphate to mannose ratio (IVW: OR:0.870, 95% CI: 0.818-0.926, p = 1.29 × 10-5, FDR = 0.008). Additionally, metabolic pathway analysis highlighted six significant pathways associated with T2DM development: Valine, leucine and isoleucine biosynthesis, Phenylalanine metabolism, Glycerophospholipid metabolism, Alpha-Linolenic acid metabolism, Sphingolipid metabolism, and Alanine, aspartate, and glutamate metabolism. Conclusion This study identifies both protective and risk-associated metabolites that play a causal role in the development of T2DM. By integrating genomics and metabolomics, it presents novel insights into the pathogenesis of T2DM. These findings hold potential implications for early screening, preventive measures, and treatment strategies for T2DM.
Collapse
Affiliation(s)
- Jixin Li
- Xiyuan Hospital of the China Academy of Chinese Medical Sciences, Beijing, China
| | - Wenru Wang
- Xiyuan Hospital of the China Academy of Chinese Medical Sciences, Beijing, China
| | - Fengzhao Liu
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Linjie Qiu
- Xiyuan Hospital of the China Academy of Chinese Medical Sciences, Beijing, China
| | - Yan Ren
- Xiyuan Hospital of the China Academy of Chinese Medical Sciences, Beijing, China
| | - Meijie Li
- Xiyuan Hospital of the China Academy of Chinese Medical Sciences, Beijing, China
| | - Wenjie Li
- Xiyuan Hospital of the China Academy of Chinese Medical Sciences, Beijing, China
| | - Feng Gao
- Xiyuan Hospital of the China Academy of Chinese Medical Sciences, Beijing, China
| | - Jin Zhang
- Xiyuan Hospital of the China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
21
|
Harada M, Adam J, Covic M, Ge J, Brandmaier S, Muschet C, Huang J, Han S, Rommel M, Rotter M, Heier M, Mohney RP, Krumsiek J, Kastenmüller G, Rathmann W, Zou Z, Zukunft S, Scheerer MF, Neschen S, Adamski J, Gieger C, Peters A, Ankerst DP, Meitinger T, Alderete TL, de Angelis MH, Suhre K, Wang-Sattler R. Bidirectional modulation of TCA cycle metabolites and anaplerosis by metformin and its combination with SGLT2i. Cardiovasc Diabetol 2024; 23:199. [PMID: 38867314 PMCID: PMC11170891 DOI: 10.1186/s12933-024-02288-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 05/28/2024] [Indexed: 06/14/2024] Open
Abstract
BACKGROUND Metformin and sodium-glucose-cotransporter-2 inhibitors (SGLT2i) are cornerstone therapies for managing hyperglycemia in diabetes. However, their detailed impacts on metabolic processes, particularly within the citric acid (TCA) cycle and its anaplerotic pathways, remain unclear. This study investigates the tissue-specific metabolic effects of metformin, both as a monotherapy and in combination with SGLT2i, on the TCA cycle and associated anaplerotic reactions in both mice and humans. METHODS Metformin-specific metabolic changes were initially identified by comparing metformin-treated diabetic mice (MET) with vehicle-treated db/db mice (VG). These findings were then assessed in two human cohorts (KORA and QBB) and a longitudinal KORA study of metformin-naïve patients with Type 2 Diabetes (T2D). We also compared MET with db/db mice on combination therapy (SGLT2i + MET). Metabolic profiling analyzed 716 metabolites from plasma, liver, and kidney tissues post-treatment, using linear regression and Bonferroni correction for statistical analysis, complemented by pathway analyses to explore the pathophysiological implications. RESULTS Metformin monotherapy significantly upregulated TCA cycle intermediates such as malate, fumarate, and α-ketoglutarate (α-KG) in plasma, and anaplerotic substrates including hepatic glutamate and renal 2-hydroxyglutarate (2-HG) in diabetic mice. Downregulated hepatic taurine was also observed. The addition of SGLT2i, however, reversed these effects, such as downregulating circulating malate and α-KG, and hepatic glutamate and renal 2-HG, but upregulated hepatic taurine. In human T2D patients on metformin therapy, significant systemic alterations in metabolites were observed, including increased malate but decreased citrulline. The bidirectional modulation of TCA cycle intermediates in mice influenced key anaplerotic pathways linked to glutaminolysis, tumorigenesis, immune regulation, and antioxidative responses. CONCLUSION This study elucidates the specific metabolic consequences of metformin and SGLT2i on the TCA cycle, reflecting potential impacts on the immune system. Metformin shows promise for its anti-inflammatory properties, while the addition of SGLT2i may provide liver protection in conditions like metabolic dysfunction-associated steatotic liver disease (MASLD). These observations underscore the importance of personalized treatment strategies.
Collapse
Affiliation(s)
- Makoto Harada
- Institute of Translational Genomics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Jonathan Adam
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Research Unit of Molecular Epidemiology, Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Marcela Covic
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Research Unit of Molecular Epidemiology, Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Jianhong Ge
- Institute of Translational Genomics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Stefan Brandmaier
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Research Unit of Molecular Epidemiology, Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Caroline Muschet
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Jialing Huang
- Institute of Translational Genomics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Siyu Han
- Institute of Translational Genomics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Martina Rommel
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Research Unit of Molecular Epidemiology, Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Markus Rotter
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Research Unit of Molecular Epidemiology, Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Margit Heier
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- KORA Study Centre, University Hospital of Augsburg, Augsburg, Germany
| | | | - Jan Krumsiek
- Institute of Computational Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Gabi Kastenmüller
- Institute of Computational Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Wolfgang Rathmann
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Institute for Biometrics and Epidemiology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany
| | - Zhongmei Zou
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Sven Zukunft
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Markus F Scheerer
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Susanne Neschen
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Jerzy Adamski
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Christian Gieger
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Research Unit of Molecular Epidemiology, Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Annette Peters
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Institute for Medical Information Processing, Biometry, and Epidemiology, Pettenkofer School of Public Health, Ludwig Maximilian University of Munich (LMU), Munich, Germany
| | - Donna P Ankerst
- Departments of Mathematics and Life Science Systems, Technical University of Munich (TUM), Garching, Germany
| | - Thomas Meitinger
- Institute of Human Genetics, Klinikum Rechts der Isar, TUM, Munich, Germany
| | - Tanya L Alderete
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, USA
| | - Martin Hrabe de Angelis
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Chair of Experimental Genetics, TUM School of Life Sciences, TUM, Freising, Germany
| | - Karsten Suhre
- Department of Physiology and Biophysics, Weill Cornell Medicine - Qatar, Education City - Qatar Foundation, Doha, Qatar
| | - Rui Wang-Sattler
- Institute of Translational Genomics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany.
- German Center for Diabetes Research (DZD), Neuherberg, Germany.
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany.
- Research Unit of Molecular Epidemiology, Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany.
| |
Collapse
|
22
|
Chen J, Wang S, Guo F, Gong Y, Chen T, Shaw C, Jiang R, Huang F, Lin D. 1H-NMR-based metabolomics reveals the preventive effect of Enteromorpha prolifera polysaccharides on diabetes in Zucker diabetic fatty rats. Food Sci Nutr 2024; 12:4049-4062. [PMID: 38873458 PMCID: PMC11167149 DOI: 10.1002/fsn3.4061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 02/02/2024] [Accepted: 02/19/2024] [Indexed: 06/15/2024] Open
Abstract
The primary objective of this investigation was to explore the beneficial impacts of Enteromorpha prolifera polysaccharide (EP) on dysglycemia in Zucker diabetic fatty (ZDF) rats, while also shedding light on its potential mechanism using 1H-NMR-based metabolomics. The results demonstrated a noteworthy reduction in fasting blood glucose (FBG, 46.3%), fasting insulin (50.17%), glycosylated hemoglobin A1c (HbA1c, 44.1%), and homeostatic model assessment of insulin resistance (HOMA-IR, 59.75%) following EP administration, while the insulin sensitivity index (ISI, 19.6%) and homeostatic model assessment of β-cell function (HOMA-β, 2.5-fold) were significantly increased. These findings indicate that EP enhances β-cell function, increases insulin sensitivity, and improves insulin resistance caused by diabetes. Moreover, EP significantly reduced serum lipid levels, suggesting improvement of dyslipidemia. Through the analysis of serum metabolomics, 17 metabolites were found to be altered in diabetic rats, 14 of which were upregulated and 3 of which were downregulated. Notably, the administration of EP successfully reversed the abnormal levels of 9 out of the 17 metabolites. Pathway analysis further revealed that EP treatment partially restored metabolic dysfunction, with notable effects observed in valine, leucine, and isoleucine metabolism; aminoacyl-transfer RNA (tRNA) biosynthesis; and ketone body metabolism. These findings collectively indicate the potential therapeutic efficacy of EP in preventing glycemic abnormalities and improving insulin resistance. Thus, EP holds promise as a valuable treatment option for individuals with diabetes.
Collapse
Affiliation(s)
- Jie Chen
- Department of Nutrition and Food Safety, School of Public HealthFujian Medical UniversityFuzhouFujianChina
| | - Shuting Wang
- Department of Nutrition and Food Safety, School of Public HealthFujian Medical UniversityFuzhouFujianChina
| | - Fuchuan Guo
- Department of Nutrition and Food Safety, School of Public HealthFujian Medical UniversityFuzhouFujianChina
| | - Yupeng Gong
- Department of Nutrition and Food Safety, School of Public HealthFujian Medical UniversityFuzhouFujianChina
| | | | - Chris Shaw
- School of PharmacyQueen's UniversityBelfastUK
| | - Rencai Jiang
- Department of Nutrition and Food Safety, School of Public HealthFujian Medical UniversityFuzhouFujianChina
| | - Fang Huang
- Department of Nutrition and Food Safety, School of Public HealthFujian Medical UniversityFuzhouFujianChina
| | - Dai Lin
- Department of Nutrition and Food Safety, School of Public HealthFujian Medical UniversityFuzhouFujianChina
| |
Collapse
|
23
|
McGee EE, Zeleznik OA, Balasubramanian R, Hu J, Rosner BA, Wactawski-Wende J, Clish CB, Avila-Pacheco J, Willett WC, Rexrode KM, Tamimi RM, Eliassen AH. Differences in metabolomic profiles between Black and White women in the U.S.: Analyses from two prospective cohorts. Eur J Epidemiol 2024; 39:653-665. [PMID: 38703248 DOI: 10.1007/s10654-024-01111-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 02/26/2024] [Indexed: 05/06/2024]
Abstract
There is growing interest in incorporating metabolomics into public health practice. However, Black women are under-represented in many metabolomics studies. If metabolomic profiles differ between Black and White women, this under-representation may exacerbate existing Black-White health disparities. We therefore aimed to estimate metabolomic differences between Black and White women in the U.S. We leveraged data from two prospective cohorts: the Nurses' Health Study (NHS; n = 2077) and Women's Health Initiative (WHI; n = 2128). The WHI served as the replication cohort. Plasma metabolites (n = 334) were measured via liquid chromatography-tandem mass spectrometry. Observed metabolomic differences were estimated using linear regression and metabolite set enrichment analyses. Residual metabolomic differences in a hypothetical population in which the distributions of 14 risk factors were equalized across racial groups were estimated using inverse odds ratio weighting. In the NHS, Black-White differences were observed for most metabolites (75 metabolites with observed differences ≥ |0.50| standard deviations). Black women had lower average levels than White women for most metabolites (e.g., for N6, N6-dimethlylysine, mean Black-White difference = - 0.98 standard deviations; 95% CI: - 1.11, - 0.84). In metabolite set enrichment analyses, Black women had lower levels of triglycerides, phosphatidylcholines, lysophosphatidylethanolamines, phosphatidylethanolamines, and organoheterocyclic compounds, but higher levels of phosphatidylethanolamine plasmalogens, phosphatidylcholine plasmalogens, cholesteryl esters, and carnitines. In a hypothetical population in which distributions of 14 risk factors were equalized, Black-White metabolomic differences persisted. Most results replicated in the WHI (88% of 272 metabolites available for replication). Substantial differences in metabolomic profiles exist between Black and White women. Future studies should prioritize racial representation.
Collapse
Affiliation(s)
- Emma E McGee
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, 677 Huntington Avenue, Boston, MA, 02115, USA.
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA.
| | - Oana A Zeleznik
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Raji Balasubramanian
- Division of Women's Health, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Jie Hu
- Division of Women's Health, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Bernard A Rosner
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Jean Wactawski-Wende
- Department of Epidemiology and Environmental Health, School of Public Health and Health Professions, University at Buffalo, Buffalo, NY, USA
| | - Clary B Clish
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
| | - Julian Avila-Pacheco
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
| | - Walter C Willett
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, 677 Huntington Avenue, Boston, MA, 02115, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Kathryn M Rexrode
- Division of Women's Health, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Rulla M Tamimi
- Department of Population Health Sciences, Weill Cornell Medical College, New York, USA
| | - A Heather Eliassen
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, 677 Huntington Avenue, Boston, MA, 02115, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| |
Collapse
|
24
|
Zhang F, Shan S, Fu C, Guo S, Liu C, Wang S. Advanced Mass Spectrometry-Based Biomarker Identification for Metabolomics of Diabetes Mellitus and Its Complications. Molecules 2024; 29:2530. [PMID: 38893405 PMCID: PMC11173766 DOI: 10.3390/molecules29112530] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 04/15/2024] [Accepted: 04/16/2024] [Indexed: 06/21/2024] Open
Abstract
Over the years, there has been notable progress in understanding the pathogenesis and treatment modalities of diabetes and its complications, including the application of metabolomics in the study of diabetes, capturing attention from researchers worldwide. Advanced mass spectrometry, including gas chromatography-tandem mass spectrometry (GC-MS/MS), liquid chromatography-tandem mass spectrometry (LC-MS/MS), and ultra-performance liquid chromatography coupled to electrospray ionization quadrupole time-of-flight mass spectrometry (UPLC-ESI-Q-TOF-MS), etc., has significantly broadened the spectrum of detectable metabolites, even at lower concentrations. Advanced mass spectrometry has emerged as a powerful tool in diabetes research, particularly in the context of metabolomics. By leveraging the precision and sensitivity of advanced mass spectrometry techniques, researchers have unlocked a wealth of information within the metabolome. This technology has enabled the identification and quantification of potential biomarkers associated with diabetes and its complications, providing new ideas and methods for clinical diagnostics and metabolic studies. Moreover, it offers a less invasive, or even non-invasive, means of tracking disease progression, evaluating treatment efficacy, and understanding the underlying metabolic alterations in diabetes. This paper summarizes advanced mass spectrometry for the application of metabolomics in diabetes mellitus, gestational diabetes mellitus, diabetic peripheral neuropathy, diabetic retinopathy, diabetic nephropathy, diabetic encephalopathy, diabetic cardiomyopathy, and diabetic foot ulcers and organizes some of the potential biomarkers of the different complications with the aim of providing ideas and methods for subsequent in-depth metabolic research and searching for new ways of treating the disease.
Collapse
Affiliation(s)
- Feixue Zhang
- Hubei Key Laboratory of Diabetes and Angiopathy, Medicine Research Institute, Medical College, Hubei University of Science and Technology, Xianning 437100, China; (F.Z.); (C.F.); (S.G.)
| | - Shan Shan
- College of Life Science, National R&D Center for Freshwater Fish Processing, Jiangxi Normal University, Nanchang 330022, China;
| | - Chenlu Fu
- Hubei Key Laboratory of Diabetes and Angiopathy, Medicine Research Institute, Medical College, Hubei University of Science and Technology, Xianning 437100, China; (F.Z.); (C.F.); (S.G.)
- School of Pharmacy, Medical College, Hubei University of Science and Technology, Xianning 437100, China
| | - Shuang Guo
- Hubei Key Laboratory of Diabetes and Angiopathy, Medicine Research Institute, Medical College, Hubei University of Science and Technology, Xianning 437100, China; (F.Z.); (C.F.); (S.G.)
| | - Chao Liu
- Hubei Key Laboratory of Diabetes and Angiopathy, Medicine Research Institute, Medical College, Hubei University of Science and Technology, Xianning 437100, China; (F.Z.); (C.F.); (S.G.)
| | - Shuanglong Wang
- Jiangxi Key Laboratory for Mass Spectrometry and Instrumentation, East China University of Technology, Nanchang 330013, China
| |
Collapse
|
25
|
Li H, Li L, Huang QQ, Yang SY, Zou JJ, Xiao F, Xiang Q, Liu X, Yu R. Global status and trends of metabolomics in diabetes: A literature visualization knowledge graph study. World J Diabetes 2024; 15:1021-1044. [PMID: 38766424 PMCID: PMC11099375 DOI: 10.4239/wjd.v15.i5.1021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 01/28/2024] [Accepted: 03/18/2024] [Indexed: 05/10/2024] Open
Abstract
BACKGROUND Diabetes is a metabolic disease characterized by hyperglycemia, which has increased the global medical burden and is also the main cause of death in most countries. AIM To understand the knowledge structure of global development status, research focus, and future trend of the relationship between diabetes and metabolomics in the past 20 years. METHODS The articles about the relationship between diabetes and metabolomics in the Web of Science Core Collection were retrieved from 2002 to October 23, 2023, and the relevant information was analyzed using CiteSpace6.2.2R (CiteSpace), VOSviewer6.1.18 (VOSviewer), and Bibliometrix software under R language. RESULTS A total of 3123 publications were included from 2002 to 2022. In the past two decades, the number of publications and citations in this field has continued to increase. The United States, China, Germany, the United Kingdom, and other relevant funds, institutions, and authors have significantly contributed to this field. Scientific Reports and PLoS One are the journals with the most publications and the most citations. Through keyword co-occurrence and cluster analysis, the closely related keywords are "insulin resistance", "risk", "obesity", "oxidative stress", "metabolomics", "metabolites" and "biomarkers". Keyword clustering included cardiovascular disease, gut microbiota, metabonomics, diabetic nephropathy, molecular docking, gestational diabetes mellitus, oxidative stress, and insulin resistance. Burst detection analysis of keyword depicted that "Gene", "microbiota", "validation", "kidney disease", "antioxidant activity", "untargeted metabolomics", "management", and "accumulation" are knowledge frontiers in recent years. CONCLUSION The relationship between metabolomics and diabetes is receiving extensive attention. Diabetic nephropathy, diabetic cardiovascular disease, and kidney disease are key diseases for future research in this field. Gut microbiota, molecular docking, and untargeted metabolomics are key research directions in the future. Antioxidant activity, gene, validation, mass spectrometry, management, and accumulation are at the forefront of knowledge frontiers in this field.
Collapse
Affiliation(s)
- Hong Li
- College of Chinese Medicine, Hunan University of Chinese Medicine, Changsha 410208, Hunan Province, China
| | - Liu Li
- College of Chinese Medicine, Hunan University of Chinese Medicine, Changsha 410208, Hunan Province, China
| | - Qiu-Qing Huang
- College of Chinese Medicine, Hunan University of Chinese Medicine, Changsha 410208, Hunan Province, China
| | - Si-Yao Yang
- College of Chinese Medicine, Hunan University of Chinese Medicine, Changsha 410208, Hunan Province, China
| | - Jun-Ju Zou
- College of Chinese Medicine, Hunan University of Chinese Medicine, Changsha 410208, Hunan Province, China
| | - Fan Xiao
- College of International Education, Hunan University of Chinese Medicine, Changsha 410208, Hunan Province, China
| | - Qin Xiang
- Department of Science and Technology, Hunan University of Chinese Medicine, Changsha 410208, Hunan Province, China
| | - Xiu Liu
- Hunan Key Laboratory of TCM Prescription and Syndromes Translational Medicine, Hunan University of Chinese Medicine, Changsha 410208, Hunan Province, China
| | - Rong Yu
- Hunan Key Laboratory of TCM Prescription and Syndromes Translational Medicine, Hunan University of Chinese Medicine, Changsha 410208, Hunan Province, China
- College of Graduate, Hunan University of Chinese Medicine, Hunan Changsha, Hunan Province, China
| |
Collapse
|
26
|
Yang C, Ma Y, Yao M, Jiang Q, Xue J. Causal relationships between blood metabolites and diabetic retinopathy: a two-sample Mendelian randomization study. Front Endocrinol (Lausanne) 2024; 15:1383035. [PMID: 38752182 PMCID: PMC11094203 DOI: 10.3389/fendo.2024.1383035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 04/05/2024] [Indexed: 05/18/2024] Open
Abstract
Background Diabetic retinopathy (DR) is a microvascular complication of diabetes, severely affecting patients' vision and even leading to blindness. The development of DR is influenced by metabolic disturbance and genetic factors, including gene polymorphisms. The research aimed to uncover the causal relationships between blood metabolites and DR. Methods The two-sample mendelian randomization (MR) analysis was employed to estimate the causality of blood metabolites on DR. The genetic variables for exposure were obtained from the genome-wide association study (GWAS) dataset of 486 blood metabolites, while the genetic predictors for outcomes including all-stage DR (All DR), non-proliferative DR (NPDR) and proliferative DR (PDR) were derived from the FinnGen database. The primary analysis employed inverse variance weighted (IVW) method, and supplementary analyses were performed using MR-Egger, weighted median (WM), simple mode and weighted mode methods. Additionally, MR-Egger intercept test, Cochran's Q test, and leave-one-out analysis were also conducted to guarantee the accuracy and robustness of the results. Subsequently, we replicated the MR analysis using three additional datasets from the FinnGen database and conducted a meta-analysis to determine blood metabolites associated with DR. Finally, reverse MR analysis and metabolic pathway analysis were performed. Results The study identified 13 blood metabolites associated with All DR, 9 blood metabolites associated with NPDR and 12 blood metabolites associated with PDR. In summary, a total of 21 blood metabolites were identified as having potential causal relationships with DR. Additionally, we identified 4 metabolic pathways that are related to DR. Conclusion The research revealed a number of blood metabolites and metabolic pathways that are causally associated with DR, which holds significant importance for screening and prevention of DR. However, it is noteworthy that these causal relationships should be validated in larger cohorts and experiments.
Collapse
Affiliation(s)
- Chongchao Yang
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
- The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yan Ma
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
- The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Mudi Yao
- Department of Ophthalmology, The First People's Hospital, Shanghai, China
| | - Qin Jiang
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
- The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jinsong Xue
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
- The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
27
|
Shao M, Chen D, Wang Q, Guo F, Wei F, Zhang W, Gan T, Luo Y, Fan X, Du P, Liu Y, Ma X, Ren G, Song Y, Zhao Y, Qin G. Canagliflozin regulates metabolic reprogramming in diabetic kidney disease by inducing fasting-like and aestivation-like metabolic patterns. Diabetologia 2024; 67:738-754. [PMID: 38236410 DOI: 10.1007/s00125-023-06078-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 11/02/2023] [Indexed: 01/19/2024]
Abstract
AIMS/HYPOTHESIS Sodium-glucose co-transporter 2 (SGLT2) inhibitors (SGLT2i) are antihyperglycaemic drugs that protect the kidneys of individuals with type 2 diabetes mellitus. However, the underlying mechanisms mediating the renal benefits of SGLT2i are not fully understood. Considering the fuel switches that occur during therapeutic SGLT2 inhibition, we hypothesised that SGLT2i induce fasting-like and aestivation-like metabolic patterns, both of which contribute to the regulation of metabolic reprogramming in diabetic kidney disease (DKD). METHODS Untargeted and targeted metabolomics assays were performed on plasma samples from participants with type 2 diabetes and kidney disease (n=35, 11 women) receiving canagliflozin (CANA) 100 mg/day at baseline and 12 week follow-up. Next, a systematic snapshot of the effect of CANA on key metabolites and pathways in the kidney was obtained using db/db mice. Moreover, the effects of glycine supplementation in db/db mice and human proximal tubular epithelial cells (human kidney-2 [HK-2]) cells were studied. RESULTS Treatment of DKD patients with CANA for 12 weeks significantly reduced HbA1c from a median (interquartile range 25-75%) of 49.0 (44.0-57.0) mmol/mol (7.9%, [7.10-9.20%]) to 42.2 (39.7-47.7) mmol/mol (6.8%, [6.40-7.70%]), and reduced urinary albumin/creatinine ratio from 67.8 (45.9-159.0) mg/mmol to 47.0 (26.0-93.6) mg/mmol. The untargeted metabolomics assay showed downregulated glycolysis and upregulated fatty acid oxidation. The targeted metabolomics assay revealed significant upregulation of glycine. The kidneys of db/db mice undergo significant metabolic reprogramming, with changes in sugar, lipid and amino acid metabolism; CANA regulated the metabolic reprogramming in the kidneys of db/db mice. In particular, the pathways for glycine, serine and threonine metabolism, as well as the metabolite of glycine, were significantly upregulated in CANA-treated kidneys. Glycine supplementation ameliorated renal lesions in db/db mice by inhibiting food intake, improving insulin sensitivity and reducing blood glucose levels. Glycine supplementation improved apoptosis of human proximal tubule cells via the AMP-activated protein kinase (AMPK)/mammalian target of rapamycin (mTOR) pathway. CONCLUSIONS/INTERPRETATION In conclusion, our study shows that CANA ameliorates DKD by inducing fasting-like and aestivation-like metabolic patterns. Furthermore, DKD was ameliorated by glycine supplementation, and the beneficial effects of glycine were probably due to the activation of the AMPK/mTOR pathway.
Collapse
Affiliation(s)
- Mingwei Shao
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Duo Chen
- Institute of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Qingzhu Wang
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Feng Guo
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Fangyi Wei
- Institute of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Wei Zhang
- Institute of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Tian Gan
- Institute of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yuanyuan Luo
- Institute of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xunjie Fan
- Institute of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Peijie Du
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yanxia Liu
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xiaojun Ma
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Gaofei Ren
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yi Song
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yanyan Zhao
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| | - Guijun Qin
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| |
Collapse
|
28
|
Baker CL, Seo KS, Park N, Rutter JK, Thornton JA, Pruett SB, Park JY. L-arginine supplementation abrogates hypoxia-induced virulence of Staphylococcus aureus in a murine diabetic pressure wound model. mSphere 2024; 9:e0077423. [PMID: 38426801 PMCID: PMC10964415 DOI: 10.1128/msphere.00774-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 01/24/2024] [Indexed: 03/02/2024] Open
Abstract
Diabetic foot ulcers (DFUs) are the most common complications of diabetes resulting from hyperglycemia leading to ischemic hypoxic tissue and nerve damage. Staphylococcus aureus is the most frequently isolated bacteria from DFUs and causes severe necrotic infections leading to amputations with a poor 5-year survival rate. However, very little is known about the mechanisms by which S. aureus dominantly colonizes and causes severe disease in DFUs. Herein, we utilized a pressure wound model in diabetic TALLYHO/JngJ mice to reproduce ischemic hypoxic tissue damage seen in DFUs and demonstrated that anaerobic fermentative growth of S. aureus significantly increased the virulence and the severity of disease by activating two-component regulatory systems leading to expression of virulence factors. Our in vitro studies showed that supplementation of nitrate as a terminal electron acceptor promotes anaerobic respiration and suppresses the expression of S. aureus virulence factors through inactivation of two-component regulatory systems, suggesting potential therapeutic benefits by promoting anaerobic nitrate respiration. Our in vivo studies revealed that dietary supplementation of L-arginine (L-Arg) significantly attenuated the severity of disease caused by S. aureus in the pressure wound model by providing nitrate. Collectively, these findings highlight the importance of anaerobic fermentative growth in S. aureus pathogenesis and the potential of dietary L-Arg supplementation as a therapeutic to prevent severe S. aureus infection in DFUs.IMPORTANCES. aureus is the most common cause of infection in DFUs, often resulting in lower-extremity amputation with a distressingly poor 5-year survival rate. Treatment for S. aureus infections has largely remained unchanged for decades and involves tissue debridement with antibiotic therapy. With high levels of conservative treatment failure, recurrence of ulcers, and antibiotic resistance, a new approach is necessary to prevent lower-extremity amputations. Nutritional aspects of DFU treatment have largely been overlooked as there has been contradictory clinical trial evidence, but very few in vitro and in vivo modelings of nutritional treatment studies have been performed. Here we demonstrate that dietary supplementation of L-Arg in a diabetic mouse model significantly reduced duration and severity of disease caused by S. aureus. These findings suggest that L-Arg supplementation could be useful as a potential preventive measure against severe S. aureus infections in DFUs.
Collapse
Affiliation(s)
- Carol L. Baker
- Department of Comparative Biomedical Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, Mississippi, USA
| | - Keun Seok Seo
- Department of Comparative Biomedical Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, Mississippi, USA
| | - Nogi Park
- Department of Comparative Biomedical Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, Mississippi, USA
| | - Jaime K. Rutter
- Department of Comparative Biomedical Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, Mississippi, USA
| | - Justin A. Thornton
- Department of Biological Sciences, College of Arts and Sciences, Mississippi State University, Mississippi State, Mississippi, USA
| | - Stephen B. Pruett
- Department of Comparative Biomedical Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, Mississippi, USA
| | - Joo Youn Park
- Department of Comparative Biomedical Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, Mississippi, USA
| |
Collapse
|
29
|
Wang Y, He X, Xue M, Yu H, He Q, Jin J. Integrated 16S rRNA sequencing and metabolomic analysis reveals the potential protective mechanism of Germacrone on diabetic nephropathy in mice. Acta Biochim Biophys Sin (Shanghai) 2024; 56:414-426. [PMID: 38429975 PMCID: PMC10984863 DOI: 10.3724/abbs.2024021] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 11/03/2023] [Indexed: 03/03/2024] Open
Abstract
Diabetic nephropathy (DN) is a severe complication of diabetes and the leading cause of end-stage renal disease and death. Germacrone (Ger) possesses anti-inflammatory, antioxidant and anti-DN properties. However, it is unclear whether the improvement in kidney damage caused by Ger in DN mice is related to abnormal compositions and metabolites of the gut microbiota. This study generates a mouse model of DN to explore the potent therapeutic ability and mechanism of Ger in renal function by 16S rRNA sequencing and untargeted fecal metabolomics. Although there is no significant change in microbiota diversity, the structure of the gut microbiota in the DN group is quite different. Serratia_marcescens and Lactobacillus_iners are elevated in the model group but significantly decreased after Ger intervention ( P<0.05). Under the treatment of Ger, no significant differences in the diversity and richness of the gut microbiota are observed. An imbalance in the intestinal flora leads to the dysregulation of metabolites, and non-targeted metabolomics data indicate high expression of stearic acid in the DN group, and oleic acid could serve as a potential marker of the therapeutic role of Ger in the DN model. Overall, Ger improves kidney injury in diabetic mice, in part potentially by reducing the abundance of Serratia_marcescens and Lactobacillus_iners, as well as regulating the associated increase in metabolites such as oleic acid, lithocholic acid and the decrease in stearic acid. Our research expands the understanding of the relationship between the gut microbiota and metabolites in Ger-treated DN. This contributes to the usage of natural products as a therapeutic approach for the treatment of DN via microbiota regulation.
Collapse
Affiliation(s)
- Yunguang Wang
- Department of Nephrologythe First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine)Hangzhou310006China
| | - Xinxin He
- Department of Nephrologythe First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine)Hangzhou310006China
| | - Mengjiao Xue
- School of Clinical MedicineHangzhou Medical CollegeHangzhou311399China
| | - Huan Yu
- The Fourth Clinical Medical CollegeZhejiang Chinese Medical UniversityHangzhou310053China
| | - Qiang He
- Department of Nephrologythe First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine)Hangzhou310006China
| | - Juan Jin
- Department of Nephrologythe First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine)Hangzhou310006China
| |
Collapse
|
30
|
Yang Y, Wang S, Sheng C, Tan J, Chen J, Li T, Ma X, Sun H, Wang X, Zhou L. Branched-chain amino acid catabolic defect promotes α-cell proliferation via activating mTOR signaling. Mol Cell Endocrinol 2024; 582:112143. [PMID: 38158148 DOI: 10.1016/j.mce.2023.112143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 12/09/2023] [Accepted: 12/21/2023] [Indexed: 01/03/2024]
Abstract
Elevated circulating level of branched-chain amino acids (BCAAs) is closely related to the development of type 2 diabetes. However, the role of BCAA catabolism in various tissues in maintaining glucose homeostasis remains largely unknown. Pancreatic α-cells have been regarded as amino acid sensors in recent years. Therefore, we generated α-cell specific branched-chain alpha-ketoacid dehydrogenase E1α subunit (BCKDHA) knockout (BCKDHA-αKO) mice to decipher the effects of BCAA catabolism in α-cells on whole-body energy metabolism. BCKDHA-αKO mice showed normal body weight, body fat, and energy expenditure. Plasma glucagon level and glucose metabolism also remained unchanged in BCKDHA-αKO mice. Whereas, the deletion of BCKDHA led to increased α-cell number due to elevated cell proliferation in neonatal mice. In vitro, only leucine among BCAAs promoted aTC1-6 cell proliferation, which was blocked by the agonist of BCAA catabolism BT2 and the inhibitor of mTOR Rapamycin. Like Rapamycin, BT2 attenuated leucine-stimulated phosphorylation of S6 in αTC1-6 cells. Elevated phosphorylation level of S6 protein in pancreatic α-cells was also observed in BCKDHA-αKO mice. These results suggest that local accumulated leucine due to defective BCAA catabolism promotes α-cell proliferation through mTOR signaling, which is insufficient to affect glucagon secretion and whole-body glucose homeostasis.
Collapse
Affiliation(s)
- Yulin Yang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shushu Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chunxiang Sheng
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jialin Tan
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Junmin Chen
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Tianjiao Li
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoqin Ma
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haipeng Sun
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Center for Cardiovascular Diseases, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Xiao Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Libin Zhou
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
31
|
Wang M, Ou Y, Yuan XL, Zhu XF, Niu B, Kang Z, Zhang B, Ahmed A, Xing GQ, Su H. Heterogeneously elevated branched-chain/aromatic amino acids among new-onset type-2 diabetes mellitus patients are potentially skewed diabetes predictors. World J Diabetes 2024; 15:53-71. [PMID: 38313852 PMCID: PMC10835491 DOI: 10.4239/wjd.v15.i1.53] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 11/03/2023] [Accepted: 12/13/2023] [Indexed: 01/12/2024] Open
Abstract
BACKGROUND The lack of specific predictors for type-2 diabetes mellitus (T2DM) severely impacts early intervention/prevention efforts. Elevated branched-chain amino acids (BCAAs: Isoleucine, leucine, valine) and aromatic amino acids (AAAs: Tyrosine, tryptophan, phenylalanine)) show high sensitivity and specificity in predicting diabetes in animals and predict T2DM 10-19 years before T2DM onset in clinical studies. However, improvement is needed to support its clinical utility. AIM To evaluate the effects of body mass index (BMI) and sex on BCAAs/AAAs in new-onset T2DM individuals with varying body weight. METHODS Ninety-seven new-onset T2DM patients (< 12 mo) differing in BMI [normal weight (NW), n = 33, BMI = 22.23 ± 1.60; overweight, n = 42, BMI = 25.9 ± 1.07; obesity (OB), n = 22, BMI = 31.23 ± 2.31] from the First People's Hospital of Yunnan Province, Kunming, China, were studied. One-way and 2-way ANOVAs were conducted to determine the effects of BMI and sex on BCAAs/AAAs. RESULTS Fasting serum AAAs, BCAAs, glutamate, and alanine were greater and high-density lipoprotein (HDL) was lower (P < 0.05, each) in OB-T2DM patients than in NW-T2DM patients, especially in male OB-T2DM patients. Arginine, histidine, leucine, methionine, and lysine were greater in male patients than in female patients. Moreover, histidine, alanine, glutamate, lysine, valine, methionine, leucine, isoleucine, tyrosine, phenylalanine, and tryptophan were significantly correlated with abdominal adiposity, body weight and BMI, whereas isoleucine, leucine and phenylalanine were negatively correlated with HDL. CONCLUSION Heterogeneously elevated amino acids, especially BCAAs/AAAs, across new-onset T2DM patients in differing BMI categories revealed a potentially skewed prediction of T2DM development. The higher BCAA/AAA levels in obese T2DM patients would support T2DM prediction in obese individuals, whereas the lower levels of BCAAs/AAAs in NW-T2DM individuals may underestimate T2DM risk in NW individuals. This potentially skewed T2DM prediction should be considered when BCAAs/AAAs are to be used as the T2DM predictor.
Collapse
Affiliation(s)
- Min Wang
- School of Chemical Science and Technology, Yunnan University, Kunming 650091, Yunnan Province, China
| | - Yang Ou
- Department of Endocrinology, The First People’s Hospital of Yunnan Province, Kunming 650032, Yunnan Province, China
| | - Xiang-Lian Yuan
- Department of Endocrinology, The First People’s Hospital of Yunnan Province, Kunming 650032, Yunnan Province, China
| | - Xiu-Fang Zhu
- School of Chemical Science and Technology, Yunnan University, Kunming 650091, Yunnan Province, China
| | - Ben Niu
- Department of Endocrinology, The First People’s Hospital of Yunnan Province, Kunming 650032, Yunnan Province, China
| | - Zhuang Kang
- Department of Endocrinology, The First People’s Hospital of Yunnan Province, Kunming 650032, Yunnan Province, China
| | - Bing Zhang
- Clinical Laboratory, Nanchong Central Hospital & The Second Clinical Medical College of North Sichuan Medical University, Nanchong 637000, Sichuan Province, China
| | - Anwar Ahmed
- Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, United States
| | - Guo-Qiang Xing
- The Affiliated Hospital and Second Clinical Medical College, North Sichuan Medical University, Nanchong 637000, Sichuan Province, China
- Department of Research and Development, Lotus Biotech.com LLC, Gaithersburg, MD 20878, United States
| | - Heng Su
- Department of Endocrinology, The First People’s Hospital of Yunnan Province, Kunming 650032, Yunnan Province, China
| |
Collapse
|
32
|
Nogal A, Alkis T, Lee Y, Kifer D, Hu J, Murphy RA, Huang Z, Wang-Sattler R, Kastenmüler G, Linkohr B, Barrios C, Crespo M, Gieger C, Peters A, Price J, Rexrode KM, Yu B, Menni C. Predictive metabolites for incident myocardial infarction: a two-step meta-analysis of individual patient data from six cohorts comprising 7897 individuals from the COnsortium of METabolomics Studies. Cardiovasc Res 2023; 119:2743-2754. [PMID: 37706562 PMCID: PMC10757581 DOI: 10.1093/cvr/cvad147] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 06/28/2023] [Accepted: 07/18/2023] [Indexed: 09/15/2023] Open
Abstract
AIMS Myocardial infarction (MI) is a major cause of death and disability worldwide. Most metabolomics studies investigating metabolites predicting MI are limited by the participant number and/or the demographic diversity. We sought to identify biomarkers of incident MI in the COnsortium of METabolomics Studies. METHODS AND RESULTS We included 7897 individuals aged on average 66 years from six intercontinental cohorts with blood metabolomic profiling (n = 1428 metabolites, of which 168 were present in at least three cohorts with over 80% prevalence) and MI information (1373 cases). We performed a two-stage individual patient data meta-analysis. We first assessed the associations between circulating metabolites and incident MI for each cohort adjusting for traditional risk factors and then performed a fixed effect inverse variance meta-analysis to pull the results together. Finally, we conducted a pathway enrichment analysis to identify potential pathways linked to MI. On meta-analysis, 56 metabolites including 21 lipids and 17 amino acids were associated with incident MI after adjusting for multiple testing (false discovery rate < 0.05), and 10 were novel. The largest increased risk was observed for the carbohydrate mannitol/sorbitol {hazard ratio [HR] [95% confidence interval (CI)] = 1.40 [1.26-1.56], P < 0.001}, whereas the largest decrease in risk was found for glutamine [HR (95% CI) = 0.74 (0.67-0.82), P < 0.001]. Moreover, the identified metabolites were significantly enriched (corrected P < 0.05) in pathways previously linked with cardiovascular diseases, including aminoacyl-tRNA biosynthesis. CONCLUSIONS In the most comprehensive metabolomic study of incident MI to date, 10 novel metabolites were associated with MI. Metabolite profiles might help to identify high-risk individuals before disease onset. Further research is needed to fully understand the mechanisms of action and elaborate pathway findings.
Collapse
Affiliation(s)
- Ana Nogal
- Department of Twin Research, King’s College London, St Thomas’ Hospital Campus, Westminster Bridge Road, SE1 7EH London, UK
| | - Taryn Alkis
- Department of Epidemiology, Human Genetics and Environmental Sciences, University of Texas Health Science Center at Houston School of Public Health, 1200 Pressler St, Suite E407, Houston, 77030 TX, USA
| | - Yura Lee
- Department of Epidemiology, Human Genetics and Environmental Sciences, University of Texas Health Science Center at Houston School of Public Health, 1200 Pressler St, Suite E407, Houston, 77030 TX, USA
| | - Domagoj Kifer
- Faculty of Pharmacy and Biochemistry, University of Zagreb, Zagreb, Croatia
| | - Jie Hu
- Division of Women’s Health, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Rachel A Murphy
- Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
- Cancer Control Research, BC Cancer, Vancouver, BC, Canada
| | - Zhe Huang
- Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, UK
| | - Rui Wang-Sattler
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, Neuherberg, Germany
| | - Gabi Kastenmüler
- Institute of Bioinformatics and Systems Biology, Helmholtz Zentrum München, Neuherberg, Germany
| | - Birgit Linkohr
- Institute of Epidemiology, Helmholtz Zentrum München, Neuherberg, Germany
| | - Clara Barrios
- Department of Nephrology, Hospital del Mar, Institut Hospital del Mar d´Investigacions Mediques, Barcelona, Spain
| | - Marta Crespo
- Department of Nephrology, Hospital del Mar, Institut Hospital del Mar d´Investigacions Mediques, Barcelona, Spain
| | - Christian Gieger
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, Neuherberg, Germany
| | - Annette Peters
- Institute of Epidemiology, Helmholtz Zentrum München, Neuherberg, Germany
| | - Jackie Price
- Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, UK
| | - Kathryn M Rexrode
- Division of Women’s Health, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Bing Yu
- Department of Epidemiology, Human Genetics and Environmental Sciences, University of Texas Health Science Center at Houston School of Public Health, 1200 Pressler St, Suite E407, Houston, 77030 TX, USA
| | - Cristina Menni
- Department of Twin Research, King’s College London, St Thomas’ Hospital Campus, Westminster Bridge Road, SE1 7EH London, UK
| |
Collapse
|
33
|
EFSA Panel on Food Additives and Flavourings (FAF), Younes M, Aquilina G, Castle L, Degen G, Engel K, Fowler PJ, Frutos Fernandez MJ, Fürst P, Gundert‐Remy U, Gürtler R, Husøy T, Manco M, Mennes W, Moldeus P, Passamonti S, Shah R, Waalkens‐Berendsen I, Wright M, Batke M, Boon P, Bruzell E, Chipman J, Crebelli R, FitzGerald R, Fortes C, Halldorsson T, LeBlanc J, Lindtner O, Mortensen A, Ntzani E, Wallace H, Barmaz S, Civitella C, D'Angelo L, Lodi F, Laganaro M, Rincon AM, Smeraldi C, Tard A. Re-evaluation of erythritol (E 968) as a food additive. EFSA J 2023; 21:e8430. [PMID: 38125972 PMCID: PMC10731997 DOI: 10.2903/j.efsa.2023.8430] [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] [Indexed: 12/23/2023] Open
Abstract
This opinion addresses the re-evaluation of erythritol (E 968) as food additive and an application for its exemption from the laxative warning label requirement as established under Regulation (EU) No 1169/2011. Erythritol is a polyol obtained by fermentation with Moniliella pollinis BC or Moniliella megachiliensis KW3-6, followed by purifications and drying. Erythritol is readily and dose-dependently absorbed in humans and can be metabolised to erythronate to a small extent. Erythritol is then excreted unchanged in the urine. It does not raise concerns regarding genotoxicity. The dataset evaluated consisted of human interventional studies. The Panel considered that erythritol has the potential to cause diarrhoea in humans, which was considered adverse because its potential association with electrolyte and water imbalance. The lower bound of the range of no observed adverse effect levels (NOAELs) for diarrhoea of 0.5 g/kg body weight (bw) was identified as reference point. The Panel considered appropriate to set a numerical acceptable daily intake (ADI) at the level of the reference point. An ADI of 0.5 g/kg bw per day was considered by the Panel to be protective for the immediate laxative effect as well as potential chronic effects, secondary to diarrhoea. The highest mean and 95th percentile chronic exposure was in children (742 mg/kg bw per day) and adolescents (1532 mg/kg bw per day). Acute exposure was maximally 3531 mg/kg bw per meal for children at the 99th percentile. Overall, the Panel considered both dietary exposure assessments an overestimation. The Panel concluded that the exposure estimates for both acute and chronic dietary exposure to erythritol (E 968) were above the ADI, indicating that individuals with high intake may be at risk of experiencing adverse effects after single and repeated exposure. Concerning the new application, the Panel concluded that the available data do not support the proposal for exemption.
Collapse
|
34
|
Ren J, Wu M. Causal effects of genetically determined blood metabolites on multiple myeloma: a Mendelian randomization study. Sci Rep 2023; 13:18818. [PMID: 37914749 PMCID: PMC10620157 DOI: 10.1038/s41598-023-45801-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 10/24/2023] [Indexed: 11/03/2023] Open
Abstract
Previous studies have shown that metabolites play an important role in phenotypic regulation. However, the causal relationship between metabolites and multiple myeloma has not been adequately investigated. Here, we attempt to explore the causal effects of genetically determined blood metabolites on multiple myeloma. The large-scale public blood metabolites and multiple myeloma datasets from independently published genome-wide association studies (GWAS) were used to explore the causal relationship between each genetically determined blood metabolite and multiple myeloma through inverse variance weighted (IVW), weighted median, MR-Egger and mode-based estimation methods. Sensitivity tests were performed to evaluate the stability and reliability of the results by MR-Egger regression and leave-one-out methods. Metabolic pathway analysis was further explored using filtered data. Statistical analyses were all performed in R. Among 452 metabolites, ten known metabolites and three unknown metabolites had significant causal relationship with multiple myeloma (P < 0.05). Four known metabolites, 3-methyl-2-oxovalenate, oxidized bilirubin, isovalerylcarnitine and glutamine carnitine, reached statistical significance in IVW models. Metabolic pathways analysis identified four significant pathways. The occurrence of multiple myeloma may have a causal relationship with these four metabolites, and there are four metabolic pathways that are also related to the occurrence of multiple myeloma. This can provide new ideas for exploring early screening and treatment of multiple myeloma.
Collapse
Affiliation(s)
- Jialin Ren
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225009, China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, 225009, China
| | - Min Wu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225009, China.
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, 225009, China.
| |
Collapse
|
35
|
Liu Y, Wang D, Liu YP. Metabolite profiles of diabetes mellitus and response to intervention in anti-hyperglycemic drugs. Front Endocrinol (Lausanne) 2023; 14:1237934. [PMID: 38027178 PMCID: PMC10644798 DOI: 10.3389/fendo.2023.1237934] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Accepted: 10/16/2023] [Indexed: 12/01/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM) has become a major health problem, threatening the quality of life of nearly 500 million patients worldwide. As a typical multifactorial metabolic disease, T2DM involves the changes and interactions of various metabolic pathways such as carbohydrates, amino acid, and lipids. It has been suggested that metabolites are not only the endpoints of upstream biochemical processes, but also play a critical role as regulators of disease progression. For example, excess free fatty acids can lead to reduced glucose utilization in skeletal muscle and induce insulin resistance; metabolism disorder of branched-chain amino acids contributes to the accumulation of toxic metabolic intermediates, and promotes the dysfunction of β-cell mitochondria, stress signal transduction, and apoptosis. In this paper, we discuss the role of metabolites in the pathogenesis of T2DM and their potential as biomarkers. Finally, we list the effects of anti-hyperglycemic drugs on serum/plasma metabolic profiles.
Collapse
Affiliation(s)
| | | | - Yi-Ping Liu
- Provincial University Key Laboratory of Sport and Health Science, School of Physical Education and Sport Sciences, Fujian Normal University, Fuzhou, China
| |
Collapse
|
36
|
Kasai S, Kokubu D, Mizukami H, Itoh K. Mitochondrial Reactive Oxygen Species, Insulin Resistance, and Nrf2-Mediated Oxidative Stress Response-Toward an Actionable Strategy for Anti-Aging. Biomolecules 2023; 13:1544. [PMID: 37892226 PMCID: PMC10605809 DOI: 10.3390/biom13101544] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 10/12/2023] [Accepted: 10/15/2023] [Indexed: 10/29/2023] Open
Abstract
Reactive oxygen species (ROS) are produced mainly by mitochondrial respiration and function as signaling molecules in the physiological range. However, ROS production is also associated with the pathogenesis of various diseases, including insulin resistance (IR) and type 2 diabetes (T2D). This review focuses on the etiology of IR and early events, especially mitochondrial ROS (mtROS) production in insulin-sensitive tissues. Importantly, IR and/or defective adipogenesis in the white adipose tissues (WAT) is thought to increase free fatty acid and ectopic lipid deposition to develop into systemic IR. Fatty acid and ceramide accumulation mediate coenzyme Q reduction and mtROS production in IR in the skeletal muscle, while coenzyme Q synthesis downregulation is also involved in mtROS production in the WAT. Obesity-related IR is associated with the downregulation of mitochondrial catabolism of branched-chain amino acids (BCAAs) in the WAT, and the accumulation of BCAA and its metabolites as biomarkers in the blood could reliably indicate future T2D. Transcription factor NF-E2-related factor 2 (Nrf2), which regulates antioxidant enzyme expression in response to oxidative stress, is downregulated in insulin-resistant tissues. However, Nrf2 inducers, such as sulforaphane, could restore Nrf2 and target gene expression and attenuate IR in multiple tissues, including the WAT.
Collapse
Affiliation(s)
- Shuya Kasai
- Department of Stress Response Science, Center for Advanced Medical Research, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki 036-8562, Japan;
| | - Daichi Kokubu
- Department of Vegetable Life Science, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki 036-8562, Japan;
- Diet & Well-being Research Institute, KAGOME CO., LTD., 17 Nishitomiyama, Nasushiobara 329-2762, Japan
| | - Hiroki Mizukami
- Department of Pathology and Molecular Medicine, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki 036-8562, Japan;
| | - Ken Itoh
- Department of Stress Response Science, Center for Advanced Medical Research, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki 036-8562, Japan;
- Department of Vegetable Life Science, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki 036-8562, Japan;
| |
Collapse
|
37
|
Mazi TA, Stanhope KL. Elevated Erythritol: A Marker of Metabolic Dysregulation or Contributor to the Pathogenesis of Cardiometabolic Disease? Nutrients 2023; 15:4011. [PMID: 37764794 PMCID: PMC10534702 DOI: 10.3390/nu15184011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 09/08/2023] [Accepted: 09/13/2023] [Indexed: 09/29/2023] Open
Abstract
Erythritol is a non-nutritive sugar replacement that can be endogenously produced by humans. Witkowski et al. reported that elevated circulating erythritol is associated with adverse cardiovascular events in three independent cohorts, demonstrated in vitro and ex vivo that erythritol promotes platelet activation, and showed faster clotting time in mice injected with erythritol. It was concluded that erythritol fosters enhanced thrombosis. This narrative review presents additional evidence that needs to be considered when evaluating these data and conclusions. We conducted a search of all studies related to erythritol exposure with focus on those that reported vascular health outcomes. Patients with chronically elevated erythritol levels due to inborn errors of metabolism do not exhibit higher platelet activation or thrombosis risk. Most long-term studies in which animals consumed high levels of erythritol do not support its role in platelet activation and thrombosis formation. Clinical data on the effects of chronic intake of erythritol are limited. Erythritol may be merely a marker of dysregulation in the Pentose Phosphate Pathway caused by impaired glycemia. However, this suggestion and the findings of Witkowski et al. need to be further examined. Clinical trials examining the long-term effects of erythritol consumption on cardiometabolic outcomes are required to test the causality between dietary erythritol and cardiometabolic risk. Until supportive data from these trials are available, it cannot be concluded that dietary erythritol promotes platelet activation, thrombosis, and cardiometabolic risk.
Collapse
Affiliation(s)
- Tagreed A. Mazi
- Department of Community Health Sciences-Clinical Nutrition, College of Applied Medical Sciences, King Saud University, P.O. Box 10219, Riyadh 11433, Saudi Arabia
| | - Kimber L. Stanhope
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA 95616, USA;
| |
Collapse
|
38
|
Ding Y, Wang S, Lu J. Unlocking the Potential: Amino Acids' Role in Predicting and Exploring Therapeutic Avenues for Type 2 Diabetes Mellitus. Metabolites 2023; 13:1017. [PMID: 37755297 PMCID: PMC10535527 DOI: 10.3390/metabo13091017] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 09/08/2023] [Accepted: 09/13/2023] [Indexed: 09/28/2023] Open
Abstract
Diabetes mellitus, particularly type 2 diabetes mellitus (T2DM), imposes a significant global burden with adverse clinical outcomes and escalating healthcare expenditures. Early identification of biomarkers can facilitate better screening, earlier diagnosis, and the prevention of diabetes. However, current clinical predictors often fail to detect abnormalities during the prediabetic state. Emerging studies have identified specific amino acids as potential biomarkers for predicting the onset and progression of diabetes. Understanding the underlying pathophysiological mechanisms can offer valuable insights into disease prevention and therapeutic interventions. This review provides a comprehensive summary of evidence supporting the use of amino acids and metabolites as clinical biomarkers for insulin resistance and diabetes. We discuss promising combinations of amino acids, including branched-chain amino acids, aromatic amino acids, glycine, asparagine and aspartate, in the prediction of T2DM. Furthermore, we delve into the mechanisms involving various signaling pathways and the metabolism underlying the role of amino acids in disease development. Finally, we highlight the potential of targeting predictive amino acids for preventive and therapeutic interventions, aiming to inspire further clinical investigations and mitigate the progression of T2DM, particularly in the prediabetic stage.
Collapse
Affiliation(s)
- Yilan Ding
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; (Y.D.); (S.W.)
- Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Shuangyuan Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; (Y.D.); (S.W.)
- Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jieli Lu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; (Y.D.); (S.W.)
- Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| |
Collapse
|
39
|
Kumar A, Prajapati P, Singh G, Kumar D, Mishra V, Kim SC, Raorane CJ, Raj V, Kushwaha S. Salbutamol Attenuates Diabetic Skeletal Muscle Atrophy by Reducing Oxidative Stress, Myostatin/GDF-8, and Pro-Inflammatory Cytokines in Rats. Pharmaceutics 2023; 15:2101. [PMID: 37631314 PMCID: PMC10458056 DOI: 10.3390/pharmaceutics15082101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 08/02/2023] [Accepted: 08/02/2023] [Indexed: 08/27/2023] Open
Abstract
Type 2 diabetes is a metabolic disorder that leads to accelerated skeletal muscle atrophy. In this study, we aimed to evaluate the effect of salbutamol (SLB) on skeletal muscle atrophy in high-fat diet (HFD)/streptozotocin (STZ)-induced diabetic rats. Male Sprague Dawley rats were divided into four groups (n = 6): control, SLB, HFD/STZ, and HFD/STZ + SLB (6 mg/kg orally for four weeks). After the last dose of SLB, rats were assessed for muscle grip strength and muscle coordination (wire-hanging, rotarod, footprint, and actophotometer tests). Body composition was analyzed in live rats. After that, animals were sacrificed, and serum and gastrocnemius (GN) muscles were collected. Endpoints include myofibrillar protein content, muscle oxidative stress and antioxidants, serum pro-inflammatory cytokines (interleukin-1β, interleukin-2, and interleukin-6), serum muscle markers (myostatin, creatine kinase, and testosterone), histopathology, and muscle 1H NMR metabolomics. Findings showed that SLB treatment significantly improved muscle strength and muscle coordination, as well as increased lean muscle mass in diabetic rats. Increased pro-inflammatory cytokines and muscle markers (myostatin, creatine kinase) indicate muscle deterioration in diabetic rats, while SLB intervention restored the same. Also, Feret's diameter and cross-sectional area of GN muscle were increased by SLB treatment, indicating the amelioration in diabetic rat muscle. Results of muscle metabolomics exhibit that SLB treatment resulted in the restoration of perturbed metabolites, including histidine-to-tyrosine, phenylalanine-to-tyrosine, and glutamate-to-glutamine ratios and succinate, sarcosine, and 3-hydroxybutyrate (3HB) in diabetic rats. These metabolites showed a pertinent role in muscle inflammation and oxidative stress in diabetic rats. In conclusion, findings showed that salbutamol could be explored as an intervention in diabetic-associated skeletal muscle atrophy.
Collapse
Affiliation(s)
- Anand Kumar
- Department of Pharmaceutical Sciences, School of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, India; (A.K.); (P.P.); (V.M.)
| | - Priyanka Prajapati
- Department of Pharmaceutical Sciences, School of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, India; (A.K.); (P.P.); (V.M.)
| | - Gurvinder Singh
- Centre of Biomedical Research, SGPGIMS Campus, Lucknow 226014, India; (G.S.); (D.K.)
| | - Dinesh Kumar
- Centre of Biomedical Research, SGPGIMS Campus, Lucknow 226014, India; (G.S.); (D.K.)
| | - Vikas Mishra
- Department of Pharmaceutical Sciences, School of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, India; (A.K.); (P.P.); (V.M.)
| | - Seong-Cheol Kim
- School of Chemical Engineering, Yeungnam University, Gyeongsan 38541, Republic of Korea;
| | | | - Vinit Raj
- School of Chemical Engineering, Yeungnam University, Gyeongsan 38541, Republic of Korea;
| | - Sapana Kushwaha
- National Institute of Pharmaceutical Education and Research (NIPER), Raebareli, New Transit Campus, Bijnor-Sisendi Road, Lucknow 226002, India
| |
Collapse
|
40
|
Wu Y, Chen T, Wang Y, Huang M, Wang Y, Luo Z. New insight into the virulence and inflammatory response of Staphylococcus aureus strains isolated from diabetic foot ulcers. Front Cell Infect Microbiol 2023; 13:1234994. [PMID: 37577369 PMCID: PMC10416727 DOI: 10.3389/fcimb.2023.1234994] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 07/14/2023] [Indexed: 08/15/2023] Open
Abstract
Staphylococcus aureus strains isolated from diabetic foot ulcers (DFUs) have less virulence, but still cause severe infections. Furthermore, hypovirulent S. aureus strains appear to be localized in the deep tissues of diabetic foot osteomyelitis, indicating that the unique environment within DFUs affects the pathogenicity of S. aureus. In this study, the cell-free culture medium (CFCM) of S. aureus strains isolated from DFUs exhibited higher cytotoxicity to human erythrocytes than those isolated from non-diabetic patients with sepsis or wounds. Among these S. aureus strains isolated from DFUs, β-toxin negative strains have less virulence than β-toxin positive strains, but induced a higher expression of inflammatory cytokines. Our study and previous studies have shown that the synergistic effect of phenol-soluble modulin α and β-toxin contributes to the higher hemolytic activity of β-toxin positive strains. However, lysis of human erythrocytes by the CFCM of β-toxin negative strains was greatly inhibited by an autolysin inhibitor, sodium polyanethole sulfonate (SPS). A high level of glucose greatly reduced the hemolytic activity of S. aureus, but promoted the expression of interleukin-6 (IL-6) in human neutrophils. However, 5 mM glucose or glucose-6-phosphate (G6P) increased the hemolytic activity of SA118 (a β-toxin negative strain) isolated from DFUs. Additionally, patients with DFUs with growth of S. aureus had lower level of serum IL-6 than those with other bacteria, and the CFCM of S. aureus strains significantly reduced lipopolysaccharide-induced IL-6 expression in human neutrophils. Therefore, the virulence and inflammatory response of S. aureus strains isolated from DFUs are determined by the levels of glucose and its metabolites, which may explain why it is the predominant bacteria isolated from DFUs.
Collapse
Affiliation(s)
- Yuan Wu
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Ti Chen
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Yanle Wang
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Mao Huang
- Department of Laboratory Medicine, Xiangya School of Medicine, Central South University, Changsha, China
| | - Yurong Wang
- Department of Laboratory Medicine, Xiangya School of Medicine, Central South University, Changsha, China
| | - Zhen Luo
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
41
|
Mendez Garcia MF, Matsuzaki S, Batushansky A, Newhardt R, Kinter C, Jin Y, Mann SN, Stout MB, Gu H, Chiao YA, Kinter M, Humphries KM. Increased cardiac PFK-2 protects against high-fat diet-induced cardiomyopathy and mediates beneficial systemic metabolic effects. iScience 2023; 26:107131. [PMID: 37534142 PMCID: PMC10391959 DOI: 10.1016/j.isci.2023.107131] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 04/27/2023] [Accepted: 06/10/2023] [Indexed: 08/04/2023] Open
Abstract
A healthy heart adapts to changes in nutrient availability and energy demands. In metabolic diseases like type 2 diabetes (T2D), increased reliance on fatty acids for energy production contributes to mitochondrial dysfunction and cardiomyopathy. A principal regulator of cardiac metabolism is 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase (PFK-2), which is a central driver of glycolysis. We hypothesized that increasing PFK-2 activity could mitigate cardiac dysfunction induced by high-fat diet (HFD). Wild type (WT) and cardiac-specific transgenic mice expressing PFK-2 (GlycoHi) were fed a low fat or HFD for 16 weeks to induce metabolic dysfunction. Metabolic phenotypes were determined by measuring mitochondrial bioenergetics and performing targeted quantitative proteomic and metabolomic analysis. Increasing cardiac PFK-2 had beneficial effects on cardiac and mitochondrial function. Unexpectedly, GlycoHi mice also exhibited sex-dependent systemic protection from HFD, including increased glucose homeostasis. These findings support improving glycolysis via PFK-2 activity can mitigate mitochondrial and functional changes that occur with metabolic syndrome.
Collapse
Affiliation(s)
- Maria F. Mendez Garcia
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Satoshi Matsuzaki
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Albert Batushansky
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
- Ilse Katz Institute for Nanoscale Science & Technology, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Ryan Newhardt
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Caroline Kinter
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Yan Jin
- Center for Translational Science, Florida International University, Port St. Lucie, FL, USA
| | - Shivani N. Mann
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Michael B. Stout
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Haiwei Gu
- Center for Translational Science, Florida International University, Port St. Lucie, FL, USA
| | - Ying Ann Chiao
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Michael Kinter
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Kenneth M. Humphries
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| |
Collapse
|
42
|
Shahisavandi M, Wang K, Ghanbari M, Ahmadizar F. Exploring Metabolomic Patterns in Type 2 Diabetes Mellitus and Response to Glucose-Lowering Medications-Review. Genes (Basel) 2023; 14:1464. [PMID: 37510368 PMCID: PMC10379356 DOI: 10.3390/genes14071464] [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: 05/17/2023] [Revised: 07/13/2023] [Accepted: 07/14/2023] [Indexed: 07/30/2023] Open
Abstract
The spectrum of information related to precision medicine in diabetes generally includes clinical data, genetics, and omics-based biomarkers that can guide personalized decisions on diabetes care. Given the remarkable progress in patient risk characterization, there is particular interest in using molecular biomarkers to guide diabetes management. Metabolomics is an emerging molecular approach that helps better understand the etiology and promises the identification of novel biomarkers for complex diseases. Both targeted or untargeted metabolites extracted from cells, biofluids, or tissues can be investigated by established high-throughput platforms, like nuclear magnetic resonance (NMR) and mass spectrometry (MS) techniques. Metabolomics is proposed as a valuable tool in precision diabetes medicine to discover biomarkers for diagnosis, prognosis, and management of the progress of diabetes through personalized phenotyping and individualized drug-response monitoring. This review offers an overview of metabolomics knowledge as potential biomarkers in type 2 diabetes mellitus (T2D) diagnosis and the response to glucose-lowering medications.
Collapse
Affiliation(s)
- Mina Shahisavandi
- Department of Epidemiology, Erasmus MC University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
| | - Kan Wang
- Department of Epidemiology, Erasmus MC University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
| | - Mohsen Ghanbari
- Department of Epidemiology, Erasmus MC University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
| | - Fariba Ahmadizar
- Department of Data Science & Biostatistics, Julius Global Health, University Medical Center Utrecht, 3508 GA Utrecht, The Netherlands
| |
Collapse
|
43
|
Xiao M, Liu W, Shi X, Wu J, Shen G, Feng J. Integration of metabolomics and network pharmacology for enhancing mechanism understanding and medication combination recommendation for diabetes mellitus and diabetic nephropathy. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2023; 15:3173-3187. [PMID: 37338009 DOI: 10.1039/d3ay00560g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2023]
Abstract
With the increasing prevalence of diabetes mellitus (DM) and diabetic nephropathy (DN), effective treatment is particularly important for the recovery of patients. However, the currently approved drugs are usually tailored to clinical symptoms and no mechanism-targeted drugs are available. In this study, the combination of metabolomics and network pharmacology was applied to provide reasonable medication combination regimens to meet the different clinical needs for the targeted treatment of DM and DN. An NMR-based metabolomic strategy was applied to identify the potential urinary biomarkers of DM or/and DN, while network pharmacology was used to identify the therapy targets of DM and DN by intersecting the targets of diseases and currently approved drugs. According to the enriched signaling pathways using the potential biomarkers and the therapy targets, the specific medication combinations were recommended for the specific clinical demands in terms of hypoglycemic, hypertensive, and/or lipid-lowering. For DM, 17 potential urinary biomarkers and 12 disease-related signaling pathways were identified, and 34 combined medication regimens related to hypoglycemia, hypoglycemia, and hypertension, and hypoglycemia, hypertension, and lipid-lowering were administered. For DN, 22 potential urinary biomarkers and 12 disease-related signaling pathways were identified, and 21 combined medication regimens related to hypoglycemia, hypoglycemia, and hypertension were proposed. Molecular docking was used to verify the binding ability, docking sites, and structure of the drug molecules to target proteins. Moreover, an integrated biological information network of the drug-target-metabolite-signaling pathways was constructed to provide insights into the underlined mechanism of DM and DN as well as clinical combination therapy.
Collapse
Affiliation(s)
- Mengxiang Xiao
- Department of Electronic Science, Fujian Provincial Key Laboratory of Plasma and Magnetic Resonance, Xiamen University, 422 Siming South Road, Siming District, Xiamen, Fujian 361005, China.
| | - Wuping Liu
- Department of Electronic Science, Fujian Provincial Key Laboratory of Plasma and Magnetic Resonance, Xiamen University, 422 Siming South Road, Siming District, Xiamen, Fujian 361005, China.
| | - Xiulin Shi
- The Xiamen Diabetes Institute and Department of Endocrinology and Diabetes, The First Affiliated Hospital of Xiamen University, Xiamen 361003, China
| | - Jinxia Wu
- Department of Electronic Science, Fujian Provincial Key Laboratory of Plasma and Magnetic Resonance, Xiamen University, 422 Siming South Road, Siming District, Xiamen, Fujian 361005, China.
| | - Guiping Shen
- Department of Electronic Science, Fujian Provincial Key Laboratory of Plasma and Magnetic Resonance, Xiamen University, 422 Siming South Road, Siming District, Xiamen, Fujian 361005, China.
| | - Jianghua Feng
- Department of Electronic Science, Fujian Provincial Key Laboratory of Plasma and Magnetic Resonance, Xiamen University, 422 Siming South Road, Siming District, Xiamen, Fujian 361005, China.
| |
Collapse
|
44
|
Ortiz SR, Field MS. Sucrose Intake Elevates Erythritol in Plasma and Urine in Male Mice. J Nutr 2023; 153:1889-1902. [PMID: 37245661 DOI: 10.1016/j.tjnut.2023.05.022] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 05/15/2023] [Accepted: 05/23/2023] [Indexed: 05/30/2023] Open
Abstract
BACKGROUND Elevated serum erythritol concentration is a predictive biomarker of diabetes and cardiovascular incidence and complications. Erythritol is synthesized endogenously from glucose, but little is known regarding the origin of elevated circulating erythritol in vivo. OBJECTIVES In vitro evidence indicates that intracellular erythritol is elevated by high-glucose cell culture conditions and that final step of erythritol synthesis is catalyzed by the enzymes sorbitol dehydrogenase (SORD) and alcohol dehydrogenase (ADH) 1. The purpose of this study was to determine whether dietary intake and/or diet-induced obesity affect erythritol synthesis in mice and whether this relationship is modified by the loss of the enzymes SORD or ADH1. METHODS First, 8-wk-old male Sord+/+, Sord-/-, Adh1+/+, and Adh1-/- mice were fed either low-fat diet (LFD) with 10% fat-derived calories or diet-induced obesity high-fat diet (HFD) with 60% fat-derived calories for 8 wk. Plasma and tissue erythritol concentrations were measured using gas chromatography-mass spectrometry. Second, male wild-type 8-wk-old C57BL/6J mice were fed LFD or HFD with plain drinking water or 30% sucrose water for 8 wk. Blood glucose and plasma and urinary erythritol concentrations were measured in nonfasted and fasted samples. Tissue erythritol was measured after killing. Finally, male Sord+/+ and Sord-/- mice were fed LFD with 30% sucrose water for 2 wk; then, nonfasted plasma, urine, and tissue erythritol concentrations were quantified. RESULTS Plasma and tissue erythritol concentrations were not affected by loss of Sord or Adh1 in mice fed LFD or HFD. In wild-type mice, consumption of 30% sucrose water significantly elevated plasma and urinary erythritol concentrations on both LFD-fed and HFD-fed mice compared with that of plain water. Sord genotype did not affect plasma or urinary erythritol concentration in response to sucrose feeding, but Sord-/- mice had reduced kidney erythritol content compared with wild-type littermates in response to sucrose. CONCLUSIONS Sucrose intake, not HFD, elevates erythritol synthesis and excretion in mice. Loss of ADH1 or SORD does not significantly affect erythritol concentration in mice.
Collapse
Affiliation(s)
- Semira R Ortiz
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, USA
| | - Martha S Field
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, USA.
| |
Collapse
|
45
|
Hotea I, Sirbu C, Plotuna AM, Tîrziu E, Badea C, Berbecea A, Dragomirescu M, Radulov I. Integrating (Nutri-)Metabolomics into the One Health Tendency-The Key for Personalized Medicine Advancement. Metabolites 2023; 13:800. [PMID: 37512507 PMCID: PMC10384896 DOI: 10.3390/metabo13070800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 06/25/2023] [Accepted: 06/26/2023] [Indexed: 07/30/2023] Open
Abstract
Metabolomics is an advanced technology, still under development, with multiple research applications, especially in the field of health. Individual metabolic profiles, the functionality of the body, as well as its interaction with the environment, can be established using this technology. The body's response to various external factors, including the food consumed and the nutrients it contains, has increased researchers' interest in nutrimetabolomics. Establishing correlations between diet and the occurrence of various diseases, or even the development of personalized nutrition plans, could contribute to advances in precision medicine. The interdependence between humans, animals, and the environment is of particular importance today, with the dramatic emergence and spread of zoonotic diseases, food, water and soil contamination, and the degradation of resources and habitats. All these events have led to an increase in risk factors for functional diseases, burdening global health. Thus, this study aimed to highlight the importance of metabolomics, and, in particular, nutrimetabolomics, as a technical solution for a holistic, collaborative, and precise approach for the advancement of the One Health strategy.
Collapse
Affiliation(s)
- Ionela Hotea
- Faculty of Veterinary Medicine, University of Life Sciences "King Mihai I" from Timisoara, Calea Aradului, No. 119, 300645 Timisoara, Romania
| | - Catalin Sirbu
- Faculty of Veterinary Medicine, University of Life Sciences "King Mihai I" from Timisoara, Calea Aradului, No. 119, 300645 Timisoara, Romania
| | - Ana-Maria Plotuna
- Faculty of Veterinary Medicine, University of Life Sciences "King Mihai I" from Timisoara, Calea Aradului, No. 119, 300645 Timisoara, Romania
| | - Emil Tîrziu
- Faculty of Veterinary Medicine, University of Life Sciences "King Mihai I" from Timisoara, Calea Aradului, No. 119, 300645 Timisoara, Romania
| | - Corina Badea
- Faculty of Veterinary Medicine, University of Life Sciences "King Mihai I" from Timisoara, Calea Aradului, No. 119, 300645 Timisoara, Romania
| | - Adina Berbecea
- Faculty of Agriculture, University of Life Sciences "King Mihai I" from Timisoara, Calea Aradului, No. 119, 300645 Timisoara, Romania
| | - Monica Dragomirescu
- Faculty of Bioengineering of Animal Resources, University of Life Sciences "King Mihai I" from Timisoara, Calea Aradului, No. 119, 300645 Timisoara, Romania
| | - Isidora Radulov
- Faculty of Agriculture, University of Life Sciences "King Mihai I" from Timisoara, Calea Aradului, No. 119, 300645 Timisoara, Romania
| |
Collapse
|
46
|
Chen T, Xu H, Yao X, Luo Z. Role of sodium pyruvate in maintaining the survival and cytotoxicity of Staphylococcus aureus under high glucose conditions. Front Microbiol 2023; 14:1209358. [PMID: 37405167 PMCID: PMC10315490 DOI: 10.3389/fmicb.2023.1209358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 06/02/2023] [Indexed: 07/06/2023] Open
Abstract
Glucose is a crucial carbon source for the growth of Staphylococcus aureus, but an excess of glucose is detrimental and even leads to cell death. Pyruvate, the central metabolite of glycolysis, has been shown to have anti-inflammatory and antioxidant properties. This study aimed to investigate the protective effect of pyruvate on S. aureus under high glucose conditions. Sodium pyruvate greatly increased the cytotoxicity of S. aureus strain BAA-1717 to human erythrocytes and neutrophils in vitro. However, the cytotoxicity and survival of S. aureus were significantly reduced by high glucose, which was restored to normal levels by the addition of sodium pyruvate. The expression of hlg and lukS in S. aureus was higher in the LB-GP cultures than that in LB-G cultures, but there was no significant difference in cytotoxicity between LB-GP and LB-G cultures. Furthermore, the hemolytic activity of S. aureus supernatants could be inhibited by the cell-free culture medium (CFCM) of LB-G cultures, suggesting that high levels of extracellular proteases were presence in the CFCM of LB-G cultures, resulting in degradation of the hemolytic factors. The expression of sarA, which negatively regulates extracellular protease secretion, was higher in LB-GP cultures than that in LB-G cultures. Additionally, sodium pyruvate increased acetate production in S. aureus, which helps maintain cell viability under acidic environment. In conclusion, pyruvate plays an important role in the survival and cytotoxicity of S. aureus under high glucose conditions. This finding may aid in the development of effective treatments for diabetic foot infections.
Collapse
Affiliation(s)
- Ti Chen
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Huan Xu
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Xiaoyan Yao
- Department of Medical Laboratory Science, School of Medicine, Hunan Normal University, Changsha, China
| | - Zhen Luo
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
47
|
Bermingham KM, Mazidi M, Franks PW, Maher T, Valdes AM, Linenberg I, Wolf J, Hadjigeorgiou G, Spector TD, Menni C, Ordovas JM, Berry SE, Hall WL. Characterisation of Fasting and Postprandial NMR Metabolites: Insights from the ZOE PREDICT 1 Study. Nutrients 2023; 15:nu15112638. [PMID: 37299601 DOI: 10.3390/nu15112638] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 05/12/2023] [Accepted: 05/18/2023] [Indexed: 06/12/2023] Open
Abstract
BACKGROUND Postprandial metabolomic profiles and their inter-individual variability are not well characterised. Here, we describe postprandial metabolite changes, their correlations with fasting values and their inter- and intra-individual variability, following a standardised meal in the ZOE PREDICT 1 cohort. METHODS In the ZOE PREDICT 1 study (n = 1002 (NCT03479866)), 250 metabolites, mainly lipids, were measured by a Nightingale NMR panel in fasting and postprandial (4 and 6 h after a 3.7 MJ mixed nutrient meal, with a second 2.2 MJ mixed nutrient meal at 4 h) serum samples. For each metabolite, inter- and intra-individual variability over time was evaluated using linear mixed modelling and intraclass correlation coefficients (ICC) were calculated. RESULTS Postprandially, 85% (of 250 metabolites) significantly changed from fasting at 6 h (47% increased, 53% decreased; Kruskal-Wallis), with 37 measures increasing by >25% and 14 increasing by >50%. The largest changes were observed in very large lipoprotein particles and ketone bodies. Seventy-one percent of circulating metabolites were strongly correlated (Spearman's rho >0.80) between fasting and postprandial timepoints, and 5% were weakly correlated (rho <0.50). The median ICC of the 250 metabolites was 0.91 (range 0.08-0.99). The lowest ICCs (ICC <0.40, 4% of measures) were found for glucose, pyruvate, ketone bodies (β-hydroxybutyrate, acetoacetate, acetate) and lactate. CONCLUSIONS In this large-scale postprandial metabolomic study, circulating metabolites were highly variable between individuals following sequential mixed meals. Findings suggest that a meal challenge may yield postprandial responses divergent from fasting measures, specifically for glycolysis, essential amino acid, ketone body and lipoprotein size metabolites.
Collapse
Affiliation(s)
- Kate M Bermingham
- Department of Nutritional Sciences, King's College London, London WC2R 2LS, UK
- Department of Twins Research and Genetic Epidemiology, King's College London, London WC2R 2LS, UK
| | - Mohsen Mazidi
- Department of Twins Research and Genetic Epidemiology, King's College London, London WC2R 2LS, UK
- Medical Research Council Population Health Research Unit, University of Oxford, Oxford OX1 3QR, UK
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK
| | - Paul W Franks
- Department of Clinical Sciences, Lund University, 21428 Malmö, Sweden
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA 02215, USA
| | - Tyler Maher
- Department of Nutritional Sciences, King's College London, London WC2R 2LS, UK
| | - Ana M Valdes
- School of Medicine, University of Nottingham, Nottingham NG5 1PB, UK
- Nottingham NIHR Biomedical Research Centre, Nottingham NG7 2UH, UK
| | - Inbar Linenberg
- Department of Nutritional Sciences, King's College London, London WC2R 2LS, UK
- ZOE Ltd., London SE1 7RW, UK
| | | | | | - Tim D Spector
- Department of Twins Research and Genetic Epidemiology, King's College London, London WC2R 2LS, UK
| | - Cristina Menni
- Department of Twins Research and Genetic Epidemiology, King's College London, London WC2R 2LS, UK
| | - Jose M Ordovas
- Jean Mayer USDA Human Nutrition Research Centre on Aging (JM-USDA-HNRCA), Tufts University, Boston, MA 02111, USA
- IMDEA Food Institute, CEI UAM + CSIC, 28049 Madrid, Spain
- Centro de Investigación Biomédica en Red-Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Sarah E Berry
- Department of Nutritional Sciences, King's College London, London WC2R 2LS, UK
| | - Wendy L Hall
- Department of Nutritional Sciences, King's College London, London WC2R 2LS, UK
| |
Collapse
|
48
|
Zhang L, Zhang H, Xie X, Tie R, Shang X, Zhao Q, Xu J, Jin L, Zhang J, Ye P. Empagliflozin ameliorates diabetic cardiomyopathy via regulated branched-chain amino acid metabolism and mTOR/p-ULK1 signaling pathway-mediated autophagy. Diabetol Metab Syndr 2023; 15:93. [PMID: 37149696 PMCID: PMC10163822 DOI: 10.1186/s13098-023-01061-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Accepted: 04/14/2023] [Indexed: 05/08/2023] Open
Abstract
BACKGROUND Empagliflozin, a sodium-glucose co-transporter 2 inhibitor (SGLT2i), has been reported to significantly reduce the risk of heart failure in multiple clinical studies. However, the underlying mechanisms remain elusive. This study aimed to investigate the effect of empagliflozin on branched-chain amino acid (BCAA) metabolism in diabetic cardiomyopathy. METHODS Thirty male 8-week KK Cg-Ay/J mice were used to study diabetic cardiomyopathy; here, 15 were used as the model group, and the remaining 15 were administered empagliflozin (3.75 mg/kg/day) by gavage daily for 16 weeks. The control group consisted of fifteen male 8-week C57BL/6J mice, whose blood glucose and body weight were measured simultaneously with the diabetic mice until 16 weeks without additional intervention. Echocardiography and histopathology were performed to evaluate cardiac structure and function. Proteomic sequencing and biogenic analysis were performed on mouse hearts. Parallel Reaction Monitoring and western blotting were performed to validate the expression levels of differentially expressed proteins. RESULTS The results showed that empagliflozin improved ventricular dilatation and ejection fraction reduction in diabetic hearts, as well as the elevation of myocardial injury biomarkers hs-cTnT and NT-proBNP. At the same time, empagliflozin alleviates myocardial inflammatory infiltration, calcification foci deposition, and fibrosis caused by diabetes. The results of the proteomics assay showed that empagliflozin could improve the metabolism of various substances, especially promoting the BCAA metabolism of diabetic hearts by up-regulating PP2Cm. Furthermore, empagliflozin could affect the mTOR/p-ULK1 signaling pathway by reducing the concentration of BCAA in diabetic hearts. When mTOR/p-ULK1 protein was inhibited, ULK1, the autophagy initiation molecule, increased. Moreover, autophagy substrate p62 and autophagy marker LC3B were significantly reduced, indicating that the autophagy activity of diabetes inhibition was reactivated. CONCLUSIONS Empagliflozin may attenuate diabetic cardiomyopathy-related myocardial injury by promoting the catabolism of BCAA and inhibiting mTOR/p-ULK1 to enhance autophagy. These findings suggest that empagliflozin could be a potential candidate drug against BCAA increase and could be used for other cardiovascular diseases with a metabolic disorder of BCAA.
Collapse
Affiliation(s)
- Lin Zhang
- Medical School of Chinese PLA, Department of Geriatric Cardiology, The Second Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Heming Zhang
- Department of Anesthesiology, The 963 Hospital of the PLA Joint Logistics Support Force, Jiamusi, China
- Department of Anesthesiology, The Second Affiliated Hospital of Air Force Medical University, Xi'an, China
| | - Xiuzhu Xie
- Medical School of Chinese PLA, Department of Geriatric Cardiology, The Second Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Ruping Tie
- Medical School of Chinese PLA, Department of Geriatric Cardiology, The Second Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Xiaolin Shang
- Department of Pharmacy, Medical Support Center of Chinese PLA General Hospital, Beijing, China
| | - Qianqian Zhao
- Medical School of Chinese PLA, Department of Cardiology, The Sixth Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Junjie Xu
- Health Service Department of the Guard Bureau of the General Office of the Central Committee of the Communist Party of China, Beijing, China
| | - Liyuan Jin
- Department of Geriatric Cardiology, The Second Medical Centre, Chinese PLA General Hospital, Beijing, China.
| | - Jinying Zhang
- Department of Basic Medicine, Medical School of Chinese PLA, Chinese PLA General Hospital, Beijing, China.
| | - Ping Ye
- Department of Geriatric Cardiology, The Second Medical Centre, Chinese PLA General Hospital, Beijing, China.
| |
Collapse
|
49
|
Su KJ, Chen XY, Gong R, Zhao Q, Hu SD, Feng MC, Li Y, Lin X, Zhang YH, Greenbaum J, Tian Q, Shen H, Xiao HM, Shen J, Deng HW. Systematic metabolomic studies identified adult adiposity biomarkers with acetylglycine associated with fat loss in vivo. Front Mol Biosci 2023; 10:1166333. [PMID: 37122566 PMCID: PMC10141311 DOI: 10.3389/fmolb.2023.1166333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 03/28/2023] [Indexed: 05/02/2023] Open
Abstract
Obesity is associated with various adverse health outcomes. Body fat (BF) distribution is recognized as an important factor of negative health consequences of obesity. Although metabolomics studies, mainly focused on body mass index (BMI) and waist circumference, have explored the biological mechanisms involved in the development of obesity, these proxy composite measures are not accurate and cannot reflect BF distribution, and thus may hinder accurate assessment of metabolic alterations and differential risk of metabolic disorders among individuals presenting adiposity differently throughout the body. Thus, the exact relations between metabolites and BF remain to be elucidated. Here, we aim to examine the associations of metabolites and metabolic pathways with BF traits which reflect BF distribution. We performed systematic untargeted serum metabolite profiling and dual-energy X-ray absorptiometry (DXA) whole body fat scan for 517 Chinese women. We jointly analyzed DXA-derived four BF phenotypes to detect cross-phenotype metabolite associations and to prioritize important metabolomic factors. Topology-based pathway analysis was used to identify important BF-related biological processes. Finally, we explored the relationships of the identified BF-related candidate metabolites with BF traits in different sex and ethnicity through two independent cohorts. Acetylglycine, the top distinguished finding, was validated for its obesity resistance effect through in vivo studies of various diet-induced obese (DIO) mice. Eighteen metabolites and fourteen pathways were discovered to be associated with BF phenotypes. Six of the metabolites were validated in varying sex and ethnicity. The obesity-resistant effects of acetylglycine were observed to be highly robust and generalizable in both human and DIO mice. These findings demonstrate the importance of metabolites associated with BF distribution patterns and several biological pathways that may contribute to obesity and obesity-related disease etiology, prevention, and intervention. Acetylglycine is highlighted as a potential therapeutic candidate for preventing excessive adiposity in future studies.
Collapse
Affiliation(s)
- Kuan-Jui Su
- Tulane Center for Biomedical Informatics and Genomics, School of Medicine, Tulane University, New Orleans, LA, United States
- Department of Biostatistics and Data Science, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, United States
| | - Xing-Ying Chen
- Shunde Hospital of Southern Medical University (The First People’s Hospital of Shunde), Foshan, China
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Rui Gong
- Tulane Center for Biomedical Informatics and Genomics, School of Medicine, Tulane University, New Orleans, LA, United States
- Shunde Hospital of Southern Medical University (The First People’s Hospital of Shunde), Foshan, China
- Department of Cadre Ward Endocrinology, Gansu Provincial Hospital, Lanzhou, China
| | - Qi Zhao
- Department of Preventive Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Shi-Di Hu
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Mei-Chen Feng
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Ye Li
- Shunde Hospital of Southern Medical University (The First People’s Hospital of Shunde), Foshan, China
| | - Xu Lin
- Tulane Center for Biomedical Informatics and Genomics, School of Medicine, Tulane University, New Orleans, LA, United States
- Shunde Hospital of Southern Medical University (The First People’s Hospital of Shunde), Foshan, China
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Yin-Hua Zhang
- Shunde Hospital of Southern Medical University (The First People’s Hospital of Shunde), Foshan, China
| | - Jonathan Greenbaum
- Tulane Center for Biomedical Informatics and Genomics, School of Medicine, Tulane University, New Orleans, LA, United States
| | - Qing Tian
- Tulane Center for Biomedical Informatics and Genomics, School of Medicine, Tulane University, New Orleans, LA, United States
| | - Hui Shen
- Tulane Center for Biomedical Informatics and Genomics, School of Medicine, Tulane University, New Orleans, LA, United States
| | - Hong-Mei Xiao
- Center of System Biology, Data Information and Reproductive Health, School of Basic Medical Science, Central South University, Changsha, China
| | - Jie Shen
- Shunde Hospital of Southern Medical University (The First People’s Hospital of Shunde), Foshan, China
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Hong-Wen Deng
- Tulane Center for Biomedical Informatics and Genomics, School of Medicine, Tulane University, New Orleans, LA, United States
| |
Collapse
|
50
|
Okut H, Lu Y, Palmer ND, Chen YDI, Taylor KD, Norris JM, Lorenzo C, Rotter JI, Langefeld CD, Wagenknecht LE, Bowden DW, Ng MCY. Metabolomic profiling of glucose homeostasis in African Americans: the Insulin Resistance Atherosclerosis Family Study (IRAS-FS). Metabolomics 2023; 19:35. [PMID: 37005925 PMCID: PMC10068644 DOI: 10.1007/s11306-023-01984-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 03/04/2023] [Indexed: 04/04/2023]
Abstract
INTRODUCTION African Americans are at increased risk for type 2 diabetes. OBJECTIVES This work aimed to examine metabolomic signature of glucose homeostasis in African Americans. METHODS We used an untargeted liquid chromatography-mass spectrometry metabolomic approach to comprehensively profile 727 plasma metabolites among 571 African Americans from the Insulin Resistance Atherosclerosis Family Study (IRAS-FS) and investigate the associations between these metabolites and both the dynamic (SI, insulin sensitivity; AIR, acute insulin response; DI, disposition index; and SG, glucose effectiveness) and basal (HOMA-IR and HOMA-B) measures of glucose homeostasis using univariate and regularized regression models. We also compared the results with our previous findings in the IRAS-FS Mexican Americans. RESULTS We confirmed increased plasma metabolite levels of branched-chain amino acids and their metabolic derivatives, 2-aminoadipate, 2-hydroxybutyrate, glutamate, arginine and its metabolic derivatives, carbohydrate metabolites, and medium- and long-chain fatty acids were associated with insulin resistance, while increased plasma metabolite levels in the glycine, serine and threonine metabolic pathway were associated with insulin sensitivity. We also observed a differential ancestral effect of glutamate on glucose homeostasis with significantly stronger effects observed in African Americans than those previously observed in Mexican Americans. CONCLUSION We extended the observations that metabolites are useful biomarkers in the identification of prediabetes in individuals at risk of type 2 diabetes in African Americans. We revealed, for the first time, differential ancestral effect of certain metabolites (i.e., glutamate) on glucose homeostasis traits. Our study highlights the need for additional comprehensive metabolomic studies in well-characterized multiethnic cohorts.
Collapse
Affiliation(s)
- Hayrettin Okut
- Center for Precision Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
- Department of Population Health, University of Kansas School of Medicine-Wichita, Wichita, KS, USA
| | - Yingchang Lu
- Division of Genetic Medicine, Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Nicholette D Palmer
- Center for Precision Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Yii-Der Ida Chen
- Department of Pediatrics, The Institute for Translational Genomics and Population Sciences, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Kent D Taylor
- Department of Pediatrics, The Institute for Translational Genomics and Population Sciences, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Jill M Norris
- Departments of Epidemiology, Colorado School of Public Health, University of Colorado Denver, Aurora, CO, USA
| | - Carlos Lorenzo
- Department of Medicine, University of Texas Health Science Center, San Antonio, TX, USA
| | - Jerome I Rotter
- Department of Pediatrics, The Institute for Translational Genomics and Population Sciences, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Carl D Langefeld
- Department of Biostatistical Sciences, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Lynne E Wagenknecht
- Division of Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Donald W Bowden
- Center for Precision Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC, USA
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Maggie C Y Ng
- Center for Precision Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA.
- Division of Genetic Medicine, Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, 37232, USA.
| |
Collapse
|