1
|
Fang X, Liu D, Zhao K, Yang S. Comment on Sprinkles et al. Advancing Diabetes Prevention and Treatment Through Choline Metabolite Research. Diabetes Care 2025; 48:e56. [PMID: 40117467 PMCID: PMC11932805 DOI: 10.2337/dc24-2133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 10/01/2024] [Indexed: 03/23/2025]
Affiliation(s)
- Xiaolu Fang
- Department of Clinical Laboratory, Xiangyang No. 1 People’s Hospital, Hubei University of Medicine, Xiangyang, China
| | - Deyang Liu
- Department of Rehabilitation Medicine, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang, China
| | - Kun Zhao
- Shanghai Jiao Tong University Affiliated Sixth People’s Hospital South Campus, Shanghai, China
| | | |
Collapse
|
2
|
He L, Gao Y, Ju C, Wang X, Zhang Y, Yu Q, Zhang L, Chen C, Duan Y. Collagen peptides alleviate hyperglycemia in mice by modulating insulin resistance, glucose metabolism and gut microbiota. Int J Biol Macromol 2025; 301:140498. [PMID: 39889980 DOI: 10.1016/j.ijbiomac.2025.140498] [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: 07/05/2024] [Revised: 01/02/2025] [Accepted: 01/28/2025] [Indexed: 02/03/2025]
Abstract
In vitro studies have demonstrated that collagen peptides (CP) inhibit the activity of dipeptidyl peptidase-IV. However, the relationship between the AMPK-mediated insulin signaling pathway and gut microbiota modulation in Type 2 Diabetes Mellitus (T2DM) remains underexplored. Current research investigated the hypoglycemic mechanisms associated with CP intervention in a T2DM mouse model induced by a high-fat diet and streptozotocin (STZ). The findings revealed that administering CP (400 mg/kg/day) for 4 weeks significantly eased symptoms such as polydipsia, polyphagia, weight loss, and organ damage in diabetic mice. Following CP intervention, diabetic mice exhibited notable reductions in blood glucose and lipid levels, as well as decreased abundance ratios of Firmicutes and Bacteroides, whereas increased short-chain fatty acid concentration in the gut microbiota and the serum GLP-1 level, accompanied by a substantial decrease in the serum insulin resistance index. Furthermore, in the livers of mice post-CP intervention, there was an upregulation of IRS1, the increase of p-AMPK/AMPK and p-GSK3β/GSK3β (P < 0.01), coupled with downregulation of PEPCK and FoxO1 expression (P < 0.05), thereby facilitating glycogen synthesis, regulation of insulin sensitivity, and inhibition of glucose production. These results provide foundational insights into the potential of collagen peptide intervention in managing and preventing T2DM.
Collapse
Affiliation(s)
- Long He
- College of Food Science and Engineering, Gansu Agricultural University, Lanzhou 730070, China.
| | - Yongfang Gao
- College of Life Science and Engineering, Lanzhou University of Technology, Lanzhou 730050, China
| | - Chaoqiang Ju
- College of Life Science and Engineering, Lanzhou University of Technology, Lanzhou 730050, China
| | - Xinyue Wang
- College of Food Science and Engineering, Gansu Agricultural University, Lanzhou 730070, China
| | - Yueyue Zhang
- College of Food Science and Engineering, Gansu Agricultural University, Lanzhou 730070, China
| | - Qunli Yu
- College of Food Science and Engineering, Gansu Agricultural University, Lanzhou 730070, China
| | - Li Zhang
- College of Food Science and Engineering, Gansu Agricultural University, Lanzhou 730070, China
| | - Cheng Chen
- College of Food Science and Engineering, Gansu Agricultural University, Lanzhou 730070, China
| | - Yufeng Duan
- College of Food Science and Engineering, Gansu Agricultural University, Lanzhou 730070, China
| |
Collapse
|
3
|
Liu J, Li F, Yang L, Luo S, Deng Y. Gut microbiota and its metabolites regulate insulin resistance: traditional Chinese medicine insights for T2DM. Front Microbiol 2025; 16:1554189. [PMID: 40177494 PMCID: PMC11963813 DOI: 10.3389/fmicb.2025.1554189] [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: 01/06/2025] [Accepted: 02/27/2025] [Indexed: 04/05/2025] Open
Abstract
The gut microbiota is closely associated with the onset and development of type 2 diabetes mellitus (T2DM), characterized by insulin resistance (IR) and chronic low-grade inflammation. However, despite the widespread use of first-line antidiabetic drugs, IR in diabetes and its complications continue to rise. The gut microbiota and its metabolic products may promote the development of T2DM by exacerbating IR. Therefore, regulating the gut microbiota has become a promising therapeutic strategy, with particular attention given to probiotics, prebiotics, synbiotics, and fecal microbiota transplantation. This review first examines the relationship between gut microbiota and IR in T2DM, summarizing the research progress of microbiota-based therapies in modulating IR. We then delve into how gut microbiota-related metabolic products contribute to IR. Finally, we summarize the research findings on the role of traditional Chinese medicine in regulating the gut microbiota and its metabolic products to improve IR. In conclusion, the gut microbiota and its metabolic products play a crucial role in the pathophysiological process of T2DM by modulating IR, offering new insights into potential therapeutic strategies for T2DM.
Collapse
Affiliation(s)
- Jing Liu
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Fuxing Li
- Ningxiang Traditional Chinese Medicine Hospital, Changsha, China
| | - Le Yang
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Shengping Luo
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Yihui Deng
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, 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
|
Mei Y, Li W, Wang B, Chen Z, Wu X, Lin Y, Wang M. Gut microbiota: an emerging target connecting polycystic ovarian syndrome and insulin resistance. Front Cell Infect Microbiol 2025; 15:1508893. [PMID: 40134784 PMCID: PMC11933006 DOI: 10.3389/fcimb.2025.1508893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 02/18/2025] [Indexed: 03/27/2025] Open
Abstract
Polycystic ovary syndrome (PCOS) is a highly heterogeneous metabolic disorder, with oligomenorrhea and hirsutism as patients' primary complaints. Hyperinsulinemia is a crucial pathophysiological mechanism in the development of PCOS, with 50-70% of patients exhibiting insulin resistance (IR). This condition not only exacerbates ovulatory dysfunction but also leads to various adverse metabolic outcomes, such as dyslipidemia and diabetes, and increases the risk of cardiovascular events both before and after menopause. Gut microbiota is a microbial community within the host that possesses significant metabolic potential and is shaped by external environmental factors, the neuro-immune network, and metabolism. Recent studies have shown that gut microbiota dysbiosis is closely related to the development and progression of PCOS. Despite the growing recognition of the potential role of gut microbiota in the pathogenesis and treatment of PCOS, its clinical application remains in its infancy. Currently, most clinical guidelines and expert consensus still emphasize traditional therapeutic approaches, such as hormonal treatments, lifestyle modifications, and insulin sensitizers. However, accumulating evidence suggests that gut microbiota may influence the metabolic and reproductive health of PCOS patients through various mechanisms. Therefore, understanding the role of gut microbiota between PCOS and IR is essential. This review describes the changes in the gut microbiota of IR-PCOS patients, examines the potential mechanisms by which the gut microbiota contributes to IR in PCOS patients, and updates the evidence supporting the gut microbiota as a potential metabolic regulatory target in IR-PCOS. In summary, gut microbiota dysbiosis may be involved in the development and progression of IR in PCOS patients, and improving gut microbiota may offer metabolic stability benefits.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Min Wang
- Department of Laboratory Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
6
|
Lu X, Ma R, Zhan J, Pan X, Liu C, Shen W, Zhang S, Zhou W, Tian Y. Thermally processed rice starch impacts glucose homeostasis in mice to different degrees via disturbing gut microbial structure and intestinal barrier function. Carbohydr Polym 2025; 348:122795. [PMID: 39562071 DOI: 10.1016/j.carbpol.2024.122795] [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: 06/26/2024] [Revised: 09/18/2024] [Accepted: 09/22/2024] [Indexed: 11/21/2024]
Abstract
Long-term intake of thermally processed starch-based foods may impact glucose homeostasis, but the consistency of the effects of various thermal treatments and the reasons are not clear. In this study, thermal treatments, especially boiling, damaged the crystal structure and inter-molecular hydrogen bonds of starch-based blends, thus decreasing the structural order and stability. These thermally treated starch-based blends increased the appetite of mice, promoted food digestion, and enhanced postprandial glucose response. Normal C57BL/6J mice were treated with boiled, baked, and fried starch-based diets for ten weeks. Compared to the baked and fried starch-based diets, the boiled starch-based diet significantly (p < 0.05) elevated random blood glucose levels and disrupted insulin homeostasis, primarily due to the remarkable decrease in gut microbial diversity. Both baked and fried starch-based diets resulted in relatively high intestinal epithelial permeability (plasma lipopolysaccharide increased by 28.67 % and 21.85 %, respectively). They adversely affected islet β-cell function and evoked glucose metabolism disorder. Overall, results demonstrate a clear connection among the thermal processing of starch-based diets, disruption of intestinal homeostasis, and adverse glucose metabolism. This study lays a theoretical foundation for the formulation of food processing strategies to mitigate the adverse effects of thermally treated food on glucose homeostasis.
Collapse
Affiliation(s)
- Xiaoxue Lu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Rongrong Ma
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Jinling Zhan
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; National Engineering Research Center of Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi 214122, China
| | - Xiaohua Pan
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Chang Liu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Wangyang Shen
- School of Food Science and Engineering, Wuhan Polytechnic University, Wuhan 430023, China
| | - Shuang Zhang
- Analysis and Testing Center, Jiangnan University, Wuxi 214122, China
| | - Weibiao Zhou
- Department of Food Science and Technology, National University of Singapore, 2 Science Drive 2, Singapore 117542, Republic of Singapore
| | - Yaoqi Tian
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China; Analysis and Testing Center, Jiangnan University, Wuxi 214122, China.
| |
Collapse
|
7
|
Yao M, Xiao Y, Sun Y, Zhang B, Ding Y, Ma Q, Liang F, Yang Z, Ge W, Liu S, Xin L, Yin J, Zhu X. Association of maternal gut microbial metabolites with gestational diabetes mellitus: evidence from an original case-control study, meta-analysis, and Mendelian randomization. Eur J Clin Nutr 2025; 79:33-41. [PMID: 39223299 DOI: 10.1038/s41430-024-01502-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 07/06/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND The associations of gut microbial metabolites, such as trimethylamine N-oxide (TMAO), its precursors, and phenylacetylglutamine (PAGln), with the risk of gestational diabetes mellitus (GDM) remain unclear. METHODS Serum samples of 201 women with GDM and 201 matched controls were collected and then targeted metabolomics was performed to examine the metabolites of interest. Multivariable conditional logistic regression was applied to investigate the relationship between metabolites and GDM. Meta-analysis was performed to combine our results and four similar articles searched from online databases, and Mendelian randomization (MR) analysis was eventually conducted to explore the causalities. RESULTS In the case-control study, after dichotomization and comparing the higher versus the lower group, the adjusted odds ratio and 95% confidence interval of choline and L-carnitine with GDM were 2.124 (1.186-3.803) and 0.293 (0.134-0.638), respectively; but neutral relationships between TMAO, betaine, and PAGln with GDM were observed. The following meta-analysis consistently revealed that L-carnitine was negatively associated with GDM. However, MR analyses showed no evidence of causalities. CONCLUSIONS Maternal levels of L-carnitine were related to the risk of GDM in both the original case-control study and meta-analysis. However, we did not observe any genetic evidence to establish a causal relationship between this metabolite and GDM.
Collapse
Affiliation(s)
- Mengxin Yao
- Suzhou Center for Disease Prevention and Control, Suzhou, China
- Department of Epidemiology and Health Statistics, Medical College of Soochow University, Suzhou, China
| | - Yue Xiao
- Department of Epidemiology and Health Statistics, Medical College of Soochow University, Suzhou, China
| | - Yanqun Sun
- Nanjing Municipal Center for Disease Control and Prevention, Nanjing, China
| | - Bing Zhang
- Department of Geriatrics, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, China
| | - Yaling Ding
- Department of Epidemiology and Health Statistics, Medical College of Soochow University, Suzhou, China
| | - Qiuping Ma
- Taicang Affiliated Hospital of Soochow University, The First People's Hospital of Taicang, 58 Changsheng Road, Suzhou, China
| | - Fei Liang
- Department of Epidemiology and Health Statistics, Medical College of Soochow University, Suzhou, China
| | - Zhuoqiao Yang
- Department of Epidemiology and Health Statistics, Medical College of Soochow University, Suzhou, China
| | - Wenxin Ge
- Department of Epidemiology and Health Statistics, Medical College of Soochow University, Suzhou, China
| | - Songliang Liu
- Taicang Affiliated Hospital of Soochow University, The First People's Hospital of Taicang, 58 Changsheng Road, Suzhou, China
| | - Lili Xin
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Major Chronic Non-communicable Diseases, Medical College of Soochow University, Suzhou, China
| | - Jieyun Yin
- Department of Epidemiology and Health Statistics, Medical College of Soochow University, Suzhou, China.
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Major Chronic Non-communicable Diseases, Medical College of Soochow University, Suzhou, China.
| | - Xiaoyan Zhu
- Suzhou Center for Disease Prevention and Control, Suzhou, China.
- School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
8
|
Wu F, Guo Y, Wang Y, Sui X, Wang H, Zhang H, Xin B, Yang C, Zhang C, Jiang S, Qu L, Feng Q, Dai Z, Shi C, Li Y. Effects of Long-Term Fasting on Gut Microbiota, Serum Metabolome, and Their Association in Male Adults. Nutrients 2024; 17:35. [PMID: 39796469 PMCID: PMC11722564 DOI: 10.3390/nu17010035] [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: 11/20/2024] [Revised: 12/11/2024] [Accepted: 12/20/2024] [Indexed: 01/13/2025] Open
Abstract
BACKGROUND Long-term fasting demonstrates greater therapeutic potential and broader application prospects in extreme environments than intermittent fasting. METHOD This pilot study of 10-day complete fasting (CF), with a small sample size of 13 volunteers, aimed to investigate the time-series impacts on gut microbiome, serum metabolome, and their interrelationships with biochemical indices. RESULTS The results show CF significantly affected gut microbiota diversity, composition, and interspecies interactions, characterized by an expansion of the Proteobacteria phylum (about six-fold) and a decrease in Bacteroidetes (about 50%) and Firmicutes (about 34%) populations. Notably, certain bacteria taxa exhibited complex interactions and strong correlations with serum metabolites implicated in energy and amino acid metabolism, with a particular focus on fatty acylcarnitines and tryptophan derivatives. A key focus of our study was the effect of Ruthenibacterium lactatiformans, which was highly increased during CF and exhibited a strong correlation with fat metabolic indicators. This bacterium was found to mitigate high-fat diet-induced obesity, glucose intolerance, dyslipidemia, and intestinal barrier dysfunction in animal experiments. These effects suggest its potential as a probiotic candidate for the amelioration of dyslipidemia and for mediating the benefits of fasting on fat metabolism. CONCLUSIONS Our pilot study suggests that alterations in gut microbiota during CF contribute to the shift of energy metabolic substrate and the establishment of a novel homeostatic state during prolonged fasting.
Collapse
Affiliation(s)
- Feng Wu
- State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University, Chongqing 200038, China
- State Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing 100094, China (Y.L.)
| | - Yaxiu Guo
- State Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing 100094, China (Y.L.)
| | - Yihua Wang
- Department of Human Microbiome, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Xiukun Sui
- State Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing 100094, China (Y.L.)
| | - Hailong Wang
- State Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing 100094, China (Y.L.)
| | - Hongyu Zhang
- State Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing 100094, China (Y.L.)
| | - Bingmu Xin
- Engineering Research Center of Human Circadian Rhythm and Sleep, Space Science and Technology Institute (Shenzhen), Shenzhen 518000, China
| | - Chao Yang
- State Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing 100094, China (Y.L.)
| | - Cheng Zhang
- Engineering Research Center of Human Circadian Rhythm and Sleep, Space Science and Technology Institute (Shenzhen), Shenzhen 518000, China
| | - Siyu Jiang
- State Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing 100094, China (Y.L.)
- School of Life Science and Technology, Harbin Institute of Technology, Harbin 150080, China
| | - Lina Qu
- State Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing 100094, China (Y.L.)
| | - Qiang Feng
- Department of Human Microbiome, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Zhongquan Dai
- State Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing 100094, China (Y.L.)
| | - Chunmeng Shi
- State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University, Chongqing 200038, China
| | - Yinghui Li
- State Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing 100094, China (Y.L.)
- School of Life Science and Technology, Harbin Institute of Technology, Harbin 150080, China
| |
Collapse
|
9
|
Nian F, Chen Y, Xia Q, Zhu C, Wu L, Lu X. Gut microbiota metabolite trimethylamine N-oxide promoted NAFLD progression by exacerbating intestinal barrier disruption and intrahepatic cellular imbalance. Int Immunopharmacol 2024; 142:113173. [PMID: 39298816 DOI: 10.1016/j.intimp.2024.113173] [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: 05/27/2024] [Revised: 09/07/2024] [Accepted: 09/11/2024] [Indexed: 09/22/2024]
Abstract
Non-alcoholic fatty liver disease (NAFLD) has become the most common chronic liver disease worldwide, with the gut microbiota and its metabolites are important regulators of its progression. Trimethylamine N-oxide (TMAO), a metabolite of the gut microbiota, has been closely associated with various metabolic diseases, but its relationship with NAFLD remains to be elucidated. In this study, we found that fecal TMAO levels correlated with NAFLD severity. Moreover, TMAO promoted lipid deposition in HepG2 fatty liver cells and exacerbated hepatic steatosis in NAFLD rats. In the colon, TMAO undermined the structure and function of the intestinal barrier at various levels, further activated the TLR4/MyD88/NF-κB pathway, and inhibited the WNT/β-catenin pathway. In the liver, TMAO induced endothelial dysfunction with capillarization of liver sinusoidal endothelial cells, while modulating macrophage polarization. In conclusion, our study suggests that gut microbiota metabolite TMAO promotes NAFLD progression by impairing the gut and liver and that targeting TMAO could be an alternative therapeutic strategy for NAFLD.
Collapse
Affiliation(s)
- Fulin Nian
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, China; Department of Gastroenterology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Yueying Chen
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qiaoyun Xia
- Department of Gastroenterology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Chen Zhu
- Department of Gastroenterology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Longyun Wu
- Department of Gastroenterology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Xiaolan Lu
- Department of Gastroenterology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China.
| |
Collapse
|
10
|
Li Z, He X, Fang Q, Yin X. Gut Microbe-Generated Metabolite Trimethylamine-N-Oxide and Ischemic Stroke. Biomolecules 2024; 14:1463. [PMID: 39595639 PMCID: PMC11591650 DOI: 10.3390/biom14111463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 11/05/2024] [Accepted: 11/08/2024] [Indexed: 11/28/2024] Open
Abstract
Trimethylamine-N-oxide (TMAO) is a gut microbiota-derived metabolite, the production of which in vivo is mainly regulated by dietary choices, gut microbiota, and the hepatic enzyme flavin monooxygenase (FMO), while its elimination occurs via the kidneys. The TMAO level is positively correlated with the risk of developing cardiovascular diseases. Recent studies have found that TMAO plays an important role in the development of ischemic stroke. In this review, we describe the relationship between TMAO and ischemic stroke risk factors (hypertension, diabetes, atrial fibrillation, atherosclerosis, thrombosis, etc.), disease risk, severity, prognostic outcomes, and recurrence and discuss the possible mechanisms by which they interact. Importantly, TMAO induces atherosclerosis and thrombosis through lipid metabolism, foam cell formation, endothelial dysfunction (via inflammation, oxidative stress, and pyroptosis), enhanced platelet hyper-reactivity, and the upregulation and activation of vascular endothelial tissue factors. Although the pathogenic mechanisms underlying TMAO's aggravation of disease severity and its effects on post-stroke neurological recovery and recurrence risk remain unclear, they may involve inflammation, astrocyte function, and pro-inflammatory monocytes. In addition, this paper provides a summary and evaluation of relevant preclinical and clinical studies on interventions regarding the gut-microbiota-dependent TMAO level to provide evidence for the prevention and treatment of ischemic stroke through the gut microbe-TMAO pathway.
Collapse
Affiliation(s)
| | | | - Qi Fang
- Department of Neurology, The First Affiliated Hospital of Soochow University, No. 899 Pinghai Road, Suzhou 215006, China; (Z.L.); (X.H.)
| | - Xulong Yin
- Department of Neurology, The First Affiliated Hospital of Soochow University, No. 899 Pinghai Road, Suzhou 215006, China; (Z.L.); (X.H.)
| |
Collapse
|
11
|
Zhou X, Zheng W, Kong W, Zeng T. Dietary patterns and diabetic microvascular complications risk: a Mendelian randomization study of European ancestry. Front Nutr 2024; 11:1429603. [PMID: 39555188 PMCID: PMC11566142 DOI: 10.3389/fnut.2024.1429603] [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: 05/08/2024] [Accepted: 10/23/2024] [Indexed: 11/19/2024] Open
Abstract
Purpose Previous observational studies about the link between dietary factors and diabetic microvascular complications (DMCs) is controversial. Thus, we systemically assessed the potential causal relationship between diet and DMCs risk using Mendelian randomization (MR) methods. Methods We used genome-wide association studies (GWAS) statistics to estimate the causal effects of 17 dietary patterns on three common DMCs in European. Summary statistics on dietary intakes were obtained from the UK biobank, and data on DMCs [diabetic retinopathy (DR), diabetic nephropathy (DN), and diabetic neuropathy (DNP)] were obtained from the FinnGen Consortium. A two-sample MR (TSMR) was conducted to explore the causal relationships of dietary habits with DMCs. In addition, multivariable MR analysis (MVMR) was performed to adjust for traditional risk factors for eating habits, and evaluated the direct or indirect effects of diet on DMCs. Results TSMR analysis revealed that salad/raw vegetable intake (odd ratio [OR]: 2.830; 95% confidence interval [CI]: 1.102-7.267; p = 0.0306) and fresh fruit intake (OR: 2.735; 95% CI: 1.622-4.611; p = 0.0002; false discovery rate [FDR] = 0.0082) increased the risk of DR, whereas cheese intake (OR: 0.742; 95% CI: 0.563-0.978; p = 0.0339) and cereal intake (OR: 0.658; 95% CI: 0.444-0.976; p = 0.0374) decreased the risk of DR. Salad/raw vegetable (OR: 6.540; 95% CI: 1.061-40.300; p = 0.0430) and fresh fruit consumption (OR: 3.573; 95% CI: 1.263-10.107; p = 0.0164) are risk factors for DN, while cereal consumption (OR: 0.380; 95% CI: 0.174-0.833; p = 0.0156) is the opposite. And genetically predicted higher pork intake increased the risk of DNP (OR: 160.971; 95% CI: 8.832-2933.974; p = 0.0006; FDR = 0.0153). The MVMR analysis revealed that cheese intake may act as an independent protective factor for DR development. Moreover, fresh fruit intake, salad/raw vegetable intake and pork intake may be independent risk factors for DR, DN and DNP, respectively. Other causal associations between dietary habits and DMCs risk may be mediated by intermediate factors. Conclusion This causal relationship study supports that specific dietary interventions may reduce the risk of DMCs.
Collapse
Affiliation(s)
- Xin Zhou
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Hubei Provincial Clinical Research Center for Diabetes and Metabolic Disorders, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Wenbin Zheng
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Hubei Provincial Clinical Research Center for Diabetes and Metabolic Disorders, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Wen Kong
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Hubei Provincial Clinical Research Center for Diabetes and Metabolic Disorders, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Tianshu Zeng
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Hubei Provincial Clinical Research Center for Diabetes and Metabolic Disorders, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
12
|
Jiang Y, Sun T, Jiang Y, Wang X, Xi Q, Dou Y, Lv H, Peng Y, Xiao S, Xu X, Liu C, Xu B, Han X, Ma H, Hu Z, Shi Z, Du J, Lin Y. Titanium exposure and gestational diabetes mellitus: associations and potential mediation by perturbation of amino acids in early pregnancy. Environ Health 2024; 23:84. [PMID: 39394610 PMCID: PMC11470715 DOI: 10.1186/s12940-024-01128-5] [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: 07/06/2024] [Accepted: 10/02/2024] [Indexed: 10/13/2024]
Abstract
BACKGROUND Several recent studies reported the potential adverse effects of titanium exposure on glucose homeostasis among the non-pregnant population, but the association of titanium exposure with gestational diabetes mellitus (GDM) is scarce. METHODS The present study of 1,449 pregnant women was conducted within the Jiangsu Birth Cohort (JBC) study in China. Urine samples were collected in the early pregnancy, and urinary titanium concentration and non-targeted metabolomics were measured. Poisson regression estimated the association of titanium exposure in the early pregnancy with subsequent risk of GDM. Multiple linear regression screened for titanium-related urine metabolites. Mediation analyses assessed the mediating effects of candidate metabolites and pathways. RESULTS As parameterized in tertiles, titanium showed positive dose-response relationship with GDM risk (P for trend = 0.008), with women at the highest tertile of titanium exposure having 30% increased risk of GDM [relative risk (RR) = 1.30 (95% CI: 1.06, 1.61)] when compared to those exposure at the first tertile level. Meanwhile, we identified the titanium-related metabolites involved in four amino acid metabolic pathways. Notably, the perturbation of the aminoacyl-tRNA biosynthesis and alanine, aspartate and glutamate metabolism mediated 27.1% and 31.0%, respectively, of the relative effect of titanium exposure on GDM. Specifically, three titanium-related metabolites, choline, creatine and L-alanine, demonstrated predominant mediation effects on the association between titanium exposure and GDM risk. CONCLUSIONS In this prospective study, we uniquely identified a correlation between early pregnancy titanium exposure and increased GDM risk. We unveiled novel insights into how perturbations in amino acid metabolism may mediate the link between titanium exposure and GDM. Notably, choline, creatine, and L-alanine emerged as key mediators influencing this association. Our findings imply that elevated titanium exposure in early pregnancy can lead to amino acid dysmetabolism, thereby elevating GDM risk.
Collapse
Affiliation(s)
- Yangqian Jiang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
- Department of Maternal, Child and Adolescent Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Tianyu Sun
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Yue Jiang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Xiaoyan Wang
- Department of Obstetrics, Gusu School, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Nanjing Medical University, Jiangsu, 215002, China
| | - Qi Xi
- Department of Obstetrics, Gusu School, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Nanjing Medical University, Jiangsu, 215002, China
| | - Yuanyan Dou
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Hong Lv
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
- State Key Laboratory of Reproductive Medicine and Offspring Health (Suzhou Centre), Gusu School, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Nanjing Medical University, Suzhou, Jiangsu, 215002, China
| | - Yuting Peng
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
- Department of Maternal, Child and Adolescent Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Shuxin Xiao
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
- Department of Maternal, Child and Adolescent Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Xin Xu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
- Department of Maternal, Child and Adolescent Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Cong Liu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Bo Xu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Xiumei Han
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Hongxia Ma
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
- State Key Laboratory of Reproductive Medicine and Offspring Health (Suzhou Centre), Gusu School, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Nanjing Medical University, Suzhou, Jiangsu, 215002, China
| | - Zhibin Hu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
- State Key Laboratory of Reproductive Medicine and Offspring Health (Suzhou Centre), Gusu School, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Nanjing Medical University, Suzhou, Jiangsu, 215002, China
| | - Zhonghua Shi
- Department of Obstetrics and Gynecology, Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, Jiangsu, 213000, China.
| | - Jiangbo Du
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China.
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China.
- State Key Laboratory of Reproductive Medicine and Offspring Health (Suzhou Centre), Gusu School, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Nanjing Medical University, Suzhou, Jiangsu, 215002, China.
| | - Yuan Lin
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China.
- Department of Maternal, Child and Adolescent Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China.
- State Key Laboratory of Reproductive Medicine and Offspring Health (Suzhou Centre), Gusu School, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Nanjing Medical University, Suzhou, Jiangsu, 215002, China.
| |
Collapse
|
13
|
Aslam MR, Perala A, Wishart AV, Hamouda RK, Elsaady E, Khan S. Therapeutic Potential of Fecal Microbiota Transplantation in Type 2 Diabetes Mellitus: A Systematic Review. Cureus 2024; 16:e70642. [PMID: 39483608 PMCID: PMC11527334 DOI: 10.7759/cureus.70642] [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: 07/31/2024] [Accepted: 09/30/2024] [Indexed: 11/03/2024] Open
Abstract
Diabetes mellitus is a chronic metabolic disease characterized by insulin resistance and hyperglycemia. It can cause various complications, which result in significant morbidity and mortality. There are multiple treatment options available to combat this disease; however, despite this, the incidence of type 2 diabetes mellitus is continuously increasing. Some promising results have shown that dysbiosis has a role in the pathogenesis of type 2 diabetes mellitus and fecal microbiota transplantation (FMT) in animals; however, the usage of FMT in humans needs further clarification and review. We explored PubMed, Popline, and Cochrane Library to identify relevant papers. Eight articles were then finalized after screening and applying eligibility criteria. These articles explored the role of the therapeutic efficacy of FMT in insulin resistance and hyperglycemia. The studies showed that the FMT had a positive impact on managing hyperglycemia and insulin resistance, which is evident in the decline of blood glucose and HBA1c levels and the rise of insulin and C-peptides. In addition, FMT also helped to control other risk factors such as hyperlipidemia and blood pressure; however, the impact on weight loss is not convincing. FMT also influenced the levels of some microbiota, which could be involved in controlling hyperglycemia and insulin resistance. Due to limited control trials and study periods and the small sample size of diabetic patients, more research is needed to explore the impact of FMT in controlling type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Muhammad Rizwan Aslam
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Alekya Perala
- Research, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Annetta V Wishart
- Research, California Institute of Behavioral Neurosciences & Psychology, Fairfield, California, USA
| | - Ranim K Hamouda
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Entesar Elsaady
- Internal Medicine/Hematology, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Safeera Khan
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| |
Collapse
|
14
|
Chen M, Gao M, Wang H, Chen Q, Liu X, Mo Q, Huang X, Ye X, Zhang D. Jingangteng capsules ameliorate liver lipid disorders in diabetic rats by regulating microflora imbalances, metabolic disorders, and farnesoid X receptor. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 132:155806. [PMID: 38876009 DOI: 10.1016/j.phymed.2024.155806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 05/30/2024] [Accepted: 06/03/2024] [Indexed: 06/16/2024]
Abstract
BACKGROUND The plant Smilax china L., also known as Jingangteng, is suspected of regulating glucose and lipid metabolism. Jingangteng capsules (JGTCs) are commonly used to treat gynecological inflammation in clinical practice. However, it is not clear whether JGTCs can regulate glucose and lipid metabolism, and the mechanism is unclear. PURPOSE To investigate the impact and mechanism of action of JGTCs on diabetes and liver lipid disorders in rats. METHODS The chemical constituents of JGTCs were examined using ultra-high-performance liquid chromatography with quadrupole time-of-flight mass spectrometry. A high-fat diet and streptozotocin-induced diabetes model was used to evaluate anti-diabetic effects by assessing blood glucose and lipid levels and liver function. The mechanism was explored using fecal 16S rRNA gene sequencing and metabolomics profiling, reverse transcription-quantiative polymerase chain reaction (RT-qPCR), and Western blot analysis. RESULTS Thirty-three components were identified in JGTCs. The serological and histomorphological assays revealed that JGTC treatment reduced levels of blood glucose and lipids, aspartate aminotransferase, alanine aminotransferase, and lipid accumulation in the liver of diabetic rats. According to 16S rDNA sequencing, JGTCs improved species richness and diversity in diabetic rats' intestinal flora and restored 22 dysregulated bacteria to control levels. Fecal metabolomics analysis showed that the altered fecal metabolites were rich in metabolites, such as histidine, taurine, low taurine, tryptophan, glycerophospholipid, and arginine. Serum metabolomics analysis indicated that serum metabolites were enriched in the metabolism of glycerophospholipids, fructose and mannose, galactose, linoleic acid, sphingolipids, histidine, valine, leucine and isoleucine biosynthesis, and tryptophan metabolism. Heatmaps revealed a strong correlation between metabolic parameters and gut microbial phylotypes. Molecular biology assays showed that JGTC treatment reversed the decreased expression of farnesoid X receptor (FXR) in the liver of diabetic rats and inhibited the expression of lipogenic genes (Srebp1c and FAS) as well as inflammation-related genes (interleukin (IL)-β, tumor necrosis factor (TNF)-α, and IL-6). Liver metabolomics analysis indicated that JGTC could significantly regulate a significant number of bile acid metabolites associated with FXR, such as glyco-beta-muricholic acid, glycocholic acid, tauro-beta-muricholic acid, and tauro-gamma-muricholic acid. CONCLUSIONS This was the first study to investigate the mechanisms of JGTCs' effects on liver lipid disorders in diabetic rats. JGTCs inhibited liver lipid accumulation and inflammatory responses in diabetic rats by affecting intestinal flora and metabolic disorders and regulating FXR-fat synthesis-related pathways to alleviate diabetic lipid disorders.
Collapse
Affiliation(s)
- Mi Chen
- Hubei Key Laboratory of Diabetes and Angiopathy, Hubei University of Science and Technology, No. 88 Xianning Avenue, Xian'an District, Xianning, Hubei Province 437100, PR China
| | - Manjun Gao
- Hubei Key Laboratory of Diabetes and Angiopathy, Hubei University of Science and Technology, No. 88 Xianning Avenue, Xian'an District, Xianning, Hubei Province 437100, PR China
| | - Hao Wang
- Hubei Key Laboratory of Resources and Chemistry of Chinese Medicine, Hubei University of Chinese Medicine, No. 16 West Huangjiahu Road, Hongshan District, Wuhan, Hubei Province 430065, PR China
| | - Qingjie Chen
- Hubei Key Laboratory of Diabetes and Angiopathy, Hubei University of Science and Technology, No. 88 Xianning Avenue, Xian'an District, Xianning, Hubei Province 437100, PR China
| | - Xiufen Liu
- Hubei Key Laboratory of Diabetes and Angiopathy, Hubei University of Science and Technology, No. 88 Xianning Avenue, Xian'an District, Xianning, Hubei Province 437100, PR China
| | - Qigui Mo
- Hubei Key Laboratory of Diabetes and Angiopathy, Hubei University of Science and Technology, No. 88 Xianning Avenue, Xian'an District, Xianning, Hubei Province 437100, PR China
| | - Xingqiong Huang
- Hubei Key Laboratory of Diabetes and Angiopathy, Hubei University of Science and Technology, No. 88 Xianning Avenue, Xian'an District, Xianning, Hubei Province 437100, PR China
| | - Xiaochuan Ye
- Hubei Key Laboratory of Resources and Chemistry of Chinese Medicine, Hubei University of Chinese Medicine, No. 16 West Huangjiahu Road, Hongshan District, Wuhan, Hubei Province 430065, PR China.
| | - Dandan Zhang
- Hubei Key Laboratory of Diabetes and Angiopathy, Hubei University of Science and Technology, No. 88 Xianning Avenue, Xian'an District, Xianning, Hubei Province 437100, PR China.
| |
Collapse
|
15
|
Abildinova GZ, Benberin VV, Vochshenkova TA, Afshar A, Mussin NM, Kaliyev AA, Zhussupova Z, Tamadon A. Global trends and collaborative networks in gut microbiota-insulin resistance research: a comprehensive bibliometric analysis (2000-2024). Front Med (Lausanne) 2024; 11:1452227. [PMID: 39211341 PMCID: PMC11358073 DOI: 10.3389/fmed.2024.1452227] [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: 06/20/2024] [Accepted: 08/01/2024] [Indexed: 09/04/2024] Open
Abstract
Background The human gut microbiota plays a crucial role in maintaining metabolic health, with substantial evidence linking its composition to insulin resistance. This study aims to analyze the global scholarly contributions on the relationship between intestinal microbiota and insulin resistance from 2000 to 2024. Methods A bibliometric analysis was conducted using data from Scopus and Web of Science Core Collection. The search strategy included terms related to "Gastrointestinal Microbiome" and "Insulin Resistance" in the title or abstract. Results The analysis of 1,884 relevant studies from 510 sources was conducted, revealing a mean citation of 51.36 per manuscript and a remarkable annual growth rate of 22.08%. The findings highlight the significant role of gut microbiota in insulin resistance, corroborating prior studies that emphasize its influence on metabolic disorders. The literature review of the current study showed key mechanisms include the regulation of short-chain fatty acids (SCFAs) and gut hormones, which are critical for glucose metabolism and inflammation regulation. The analysis also identifies "Food and Function" as the most productive journal and Nieuwdorp M. as a leading author, underscoring the collaborative nature of this research area. Conclusion The consistent increase in publications in the field of gut microbiota and insulin resistance indicates growing recognition of the gut microbiota's therapeutic potential in treating insulin resistance and related metabolic disorders. Future research should focus on standardizing methodologies and conducting large-scale clinical trials to fully realize these therapeutic possibilities.
Collapse
Affiliation(s)
- Gulshara Zh Abildinova
- Gerontology Center, Medical Center Hospital of the President’s Affairs Administration of the Republic of Kazakhstan, Astana, Kazakhstan
| | - Valeriy V. Benberin
- Gerontology Center, Medical Center Hospital of the President’s Affairs Administration of the Republic of Kazakhstan, Astana, Kazakhstan
- Corporate Foundation, Institute of Innovative and Preventive Medicine, Astana, Kazakhstan
| | - Tamara A. Vochshenkova
- Gerontology Center, Medical Center Hospital of the President’s Affairs Administration of the Republic of Kazakhstan, Astana, Kazakhstan
| | - Alireza Afshar
- Student Research Committee, Bushehr University of Medical Sciences, Bushehr, Iran
- PerciaVista R&D Co., Shiraz, Iran
| | - Nadiar M. Mussin
- Department of Surgery No. 2, West Kazakhstan Medical University, Aktobe, Kazakhstan
| | - Asset A. Kaliyev
- Department of Surgery No. 2, West Kazakhstan Medical University, Aktobe, Kazakhstan
| | - Zhanna Zhussupova
- Department of Neurology, Psychiatry and Narcology, West Kazakhstan Marat Ospanov Medical University, Aktobe, Kazakhstan
| | - Amin Tamadon
- PerciaVista R&D Co., Shiraz, Iran
- Department of Natural Sciences, West Kazakhstan Marat Ospanov Medical University, Aktobe, Kazakhstan
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
16
|
Yu X, Wang Y, Yang R, Wang Z, Wang X, Wang S, Zhang W, Dong J, Chen W, Ji F, Gao W. Trimethylamine N-oxide predicts cardiovascular events in coronary artery disease patients with diabetes mellitus: a prospective cohort study. Front Endocrinol (Lausanne) 2024; 15:1360861. [PMID: 39092284 PMCID: PMC11291261 DOI: 10.3389/fendo.2024.1360861] [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: 12/24/2023] [Accepted: 06/27/2024] [Indexed: 08/04/2024] Open
Abstract
Background Gut microbiota has significant impact on the cardio-metabolism and inflammation, and is implicated in the pathogenesis and progression of atherosclerosis. However, the long-term prospective association between trimethylamine N-oxide (TMAO) level and major adverse clinical events (MACEs) in patients with coronary artery disease (CAD) with or without diabetes mellitus (DM) habitus remains to be investigated. Methods This prospective, single-center cohort study enrolled 2090 hospitalized CAD patients confirmed by angiography at Beijing Hospital from 2017-2020. TMAO levels were performed using liquid chromatography-tandem mass spectrometry. The composite outcome of MACEs was identified by clinic visits or interviews annually. Multivariate Cox regression analysis, Kaplan-Meier analysis, and restricted cubic splines were mainly used to explore the relationship between TMAO levels and MACEs based on diabetes mellitus (DM) habitus. Results During the median follow-up period of 54 (41, 68) months, 266 (12.7%) developed MACEs. Higher TMAO levels, using the tertile cut-off value of 318.28 ng/mL, were significantly found to be positive dose-independent for developing MACEs, especially in patients with DM (HR 1.744, 95%CI 1.084-2.808, p = 0.022). Conclusions Higher levels of TMAO are significantly associated with long-term MACEs among CAD patients with DM. The combination of TMAO in patients with CAD and DM is beneficial for risk stratification and prognosis.
Collapse
Affiliation(s)
- Xue Yu
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Yijia Wang
- Fuwai Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College/National Center for Cardiovascular Diseases, Beijing, China
| | - Ruiyue Yang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, China
| | - Zhe Wang
- Department of Cardiology, China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xinyue Wang
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Siming Wang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, China
| | - Wenduo Zhang
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Jun Dong
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, China
| | - Wenxiang Chen
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, China
| | - Fusui Ji
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Wei Gao
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University; NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Peking University; Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, China
| |
Collapse
|
17
|
Kumbhare SV, Pedroso I, Joshi B, Muthukumar KM, Saravanan SK, Irudayanathan C, Kochhar GS, Dulai PS, Sinha R, Almonacid DE. Longitudinal gut microbial signals are associated with weight loss: insights from a digital therapeutics program. Front Nutr 2024; 11:1363079. [PMID: 39040930 PMCID: PMC11262244 DOI: 10.3389/fnut.2024.1363079] [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: 12/29/2023] [Accepted: 06/24/2024] [Indexed: 07/24/2024] Open
Abstract
Introduction The gut microbiome's influence on weight management has gained significant interest for its potential to support better obesity therapeutics. Patient stratification leading to personalized nutritional intervention has shown benefits over one-size-fit-all diets. However, the efficacy and impact on the gut's microbiome of personalizing weight loss diets based on individual factors remains under-investigated. Methods This study assessed the impact of Digbi Health's personalized dietary and lifestyle program on weight loss and the gut microbiome end-points in 103 individuals. Participants' weight loss patterns and gut microbiome profiles were analyzed from baseline to follow-up samples. Results Specific microbial genera, functional pathways, and communities associated with BMI changes and the program's effectiveness were identified. 80% of participants achieved weight loss. Analysis of the gut microbiome identified genera and functional pathways associated with a reduction in BMI, including Akkermansia, Christensenella, Oscillospiraceae, Alistipes, and Sutterella, short-chain fatty acid production, and degradation of simple sugars like arabinose, sucrose, and melibiose. Network analysis identified a microbiome community associated with BMI, which includes multiple taxa known for associations with BMI and obesity. Discussion The personalized dietary and lifestyle program positively impacted the gut microbiome and demonstrated significant associations between gut microbial changes and weight loss. These findings support the use of the gut microbiome as an endpoint in weight loss interventions, highlighting potential microbiome biomarkers for further research.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Gursimran S. Kochhar
- Division of Gastroenterology, Hepatology and Nutrition, Allegheny Health Network, Pittsburgh, PA, United States
| | - Parambir S. Dulai
- Division of Gastroenterology, Northwestern University, Chicago, IL, United States
| | | | | |
Collapse
|
18
|
Li J, Bai J, Yang Y, Wu Z. Low-protein diet supplemented with 1% L-glutamine improves growth performance, serum biochemistry, redox status, plasma amino acids, and alters fecal microbiota in weaned piglets. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2024; 17:144-154. [PMID: 38766517 PMCID: PMC11101948 DOI: 10.1016/j.aninu.2023.12.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 12/14/2023] [Accepted: 12/28/2023] [Indexed: 05/22/2024]
Abstract
Glutamine, one of the most abundant amino acids in the body, has been shown to exert various beneficial effects in pigs. However, knowledge regarding the role of dietary glutamine in low-protein diet-fed piglets remains scarce. The present study aimed to investigate the effects of different levels of L-glutamine on growth performance, serum biochemistry parameters, redox status, amino acids, and fecal microbiota in low-protein diet-fed piglets. A total of 128 healthy crossbred piglets (Landrace × Yorkshire) were randomly allocated into 4 groups of 4 replicate pens, with 8 piglets per pen. Piglets in the 4 groups were fed with corn and soybean meal-based low-protein diets (crude protein level, 17%) that contained 0%, 1%, 2%, and 3% L-glutamine, respectively, for 28 d. Pigs administered 1% L-glutamine had greater body weight on d 28 and average daily gain (ADG, P < 0.01), whereas a lower feed to gain ratio (F:G) from d 1 to 28 (P < 0.01), compared to the other three groups. Besides, lower body weight on d 14 and 28, ADG, average daily feed intake, and higher F:G from d 15 to 28 and d 1 to 28 were observed in response to 2% and 3% L-glutamine treatments than 0% and 1% L-glutamine treatments (P < 0.01). Moreover, 1% L-glutamine reduced serum glucose, malondialdehyde, hydrogen peroxide concentrations and inhibited aspartate aminotransferase, alanine aminotransferase, myeloperoxidase activities in low-protein diet-fed piglets on d 14, with concomitantly upregulated catalase, total superoxide dismutase activities and glutathione level (P < 0.05). However, dietary 3% L-glutamine enhanced blood urea nitrogen content in pigs on d 14 (P < 0.05). Further investigation revealed that 1% L-glutamine upregulated the serum glutamine, lysine, methionine, tyrosine, and reduced plasma valine content (P < 0.05). Additionally, 1% L-glutamine upregulated the abundance of p_75_a5, Clostridium, Lactobacillus, Prevotellaceae_Prevotella, and Gemmiger in the stool of piglets on d 14, with the Streptococcus level being concomitantly reduced (P < 0.05). Collectively, dietary 1% L-glutamine enhances the growth performance and improves serum physiochemical parameters and antioxidative capacity in low-protein diet-fed piglets at an early age, which are associated with an increased synthesis of glutathione by modulating amino acid levels, and the optimization of gut microbiota.
Collapse
Affiliation(s)
- Jun Li
- State Key Laboratory of Animal Nutrition and Feeding, Department of Companion Animal Science, China Agricultural University, Beijing 100193, China
| | - Jun Bai
- State Key Laboratory of Animal Nutrition and Feeding, Department of Companion Animal Science, China Agricultural University, Beijing 100193, China
| | - Ying Yang
- State Key Laboratory of Animal Nutrition and Feeding, Department of Companion Animal Science, China Agricultural University, Beijing 100193, China
| | - Zhenlong Wu
- State Key Laboratory of Animal Nutrition and Feeding, Department of Companion Animal Science, China Agricultural University, Beijing 100193, China
| |
Collapse
|
19
|
Babu A, Devi Rajeswari V, Ganesh V, Das S, Dhanasekaran S, Usha Rani G, Ramanathan G. Gut Microbiome and Polycystic Ovary Syndrome: Interplay of Associated Microbial-Metabolite Pathways and Therapeutic Strategies. Reprod Sci 2024; 31:1508-1520. [PMID: 38228976 DOI: 10.1007/s43032-023-01450-2] [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: 09/20/2023] [Accepted: 12/28/2023] [Indexed: 01/18/2024]
Abstract
Polycystic ovary syndrome (PCOS) is a multifaceted disease with an intricate etiology affecting reproductive-aged women. Despite attempts to unravel the pathophysiology, the molecular mechanism of PCOS remains unknown. There are no effective or suitable therapeutic strategies available to ameliorate PCOS; however, the symptoms can be managed. In recent years, a strong association has been found between the gut microbiome and PCOS, leading to the formulation of novel ideas on the genesis and pathological processes of PCOS. Further, gut microbiome dysbiosis involving microbial metabolites may trigger PCOS symptoms via many mechanistic pathways including those associated with carbohydrates, short-chain fatty acids, lipopolysaccharides, bile acids, and gut-brain axis. We present the mechanistic pathways of PCOS-related microbial metabolites and therapeutic opportunities available to treat PCOS, such as prebiotics, probiotics, and fecal microbiota therapy. In addition, the current review highlights the emerging treatment strategies available to alleviate the symptoms of PCOS.
Collapse
Affiliation(s)
- Achsha Babu
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India
| | - V Devi Rajeswari
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India
| | - V Ganesh
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India
| | - Soumik Das
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India
| | - Sivaraman Dhanasekaran
- Pandit Deendayal Energy University, Knowledge Corridor, Raisan Village, PDPU Road, Gandhinagar, Gujarat, 382426, India
| | - G Usha Rani
- Department of Obstetrics And Gynecology, Sri Ramachandra Institute of Higher Education and Research, Chennai, India
| | - Gnanasambandan Ramanathan
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India.
| |
Collapse
|
20
|
Gao Y, Wang J, Xiao Y, Yu L, Tang Q, Wang Y, Zhou J. Structure characterization of an agavin-type fructan isolated from Polygonatum cyrtonema and its effect on the modulation of the gut microbiota in vitro. Carbohydr Polym 2024; 330:121829. [PMID: 38368108 DOI: 10.1016/j.carbpol.2024.121829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 01/11/2024] [Accepted: 01/12/2024] [Indexed: 02/19/2024]
Abstract
The herbal medicine Polygonatum cyrtonema is highly regarded in China for its medicinal and dietary properties. However, further research is needed to elucidate the structure of its polysaccharide and understand how it promotes human health by modulating the gut microbiota. This study aims to investigate a homogeneous polysaccharide (PCP95-1-1) from Polygonatum cyrtonema and assess its susceptibility to digestion as well as its utilization by intestinal microbiota. The results confirmed that PCP95-1-1 is an agavin-type fructan, which possesses two fructose chains, namely β-(2 → 6) and β-(2 → 1) fructosyl-fructose, attached to the sucrose core, and has branches of β-D-Fruf residues. Moreover, PCP95-1-1 demonstrated resistance to digestion and maintained its reducing sugar content throughout the digestive system, indicating it could reach the gut without being digested. In vitro fermentation of PCP95-1-1 significantly decreased the pH value (p < 0.05) while notably increasing the production of short-chain fatty acids (SCFAs), confirming its utilization by human gut microbiota. Additionally, PCP95-1-1 exhibited a significant ability (p < 0.05) to beneficial bacteria such as Megamonas and Bifidobacterium, while reducing the presence of facultative or conditional pathogens such as Escherichia-Shigella and Klebsiella at the genus level. Consequently, PCP95-1-1 has the potential to positively influence physical well-being by modulating the gut microbiota environment and can be developed as a functional food.
Collapse
Affiliation(s)
- Ya Gao
- School of Perfume and Aroma Technology, Shanghai Institute of Technology, Shanghai 201418, China; Institute of Edible Fungi, Shanghai Academy of Agricultural Sciences, Shanghai 201403, China
| | - Jinyan Wang
- Institute of Edible Fungi, Shanghai Academy of Agricultural Sciences, Shanghai 201403, China
| | - Ying Xiao
- School of Food and Tourism, Shanghai Urban Construction Vocational College, Shanghai 201415, China.
| | - Ling Yu
- School of Perfume and Aroma Technology, Shanghai Institute of Technology, Shanghai 201418, China.
| | - Qingjiu Tang
- Institute of Edible Fungi, Shanghai Academy of Agricultural Sciences, Shanghai 201403, China
| | - Yipeng Wang
- School of Perfume and Aroma Technology, Shanghai Institute of Technology, Shanghai 201418, China
| | - Jianjin Zhou
- Sanming Academy of Agricultural Sciences, Fujian Key Laboratory of Crop Genetic Improvement and Innovative Utilization for Mountain Area, Sanming 365051, China
| |
Collapse
|
21
|
Li S, Yang D, Zhou X, Chen L, Liu L, Lin R, Li X, Liu Y, Qiu H, Cao H, Liu J, Cheng Q. Neurological and metabolic related pathophysiologies and treatment of comorbid diabetes with depression. CNS Neurosci Ther 2024; 30:e14497. [PMID: 37927197 PMCID: PMC11017426 DOI: 10.1111/cns.14497] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 09/26/2023] [Accepted: 10/03/2023] [Indexed: 11/07/2023] Open
Abstract
BACKGROUND The comorbidity between diabetes mellitus and depression was revealed, and diabetes mellitus increased the prevalence of depressive disorder, which ranked 13th in the leading causes of disability-adjusted life-years. Insulin resistance, which is common in diabetes mellitus, has increased the risk of depressive symptoms in both humans and animals. However, the mechanisms behind the comorbidity are multi-factorial and complicated. There is still no causal chain to explain the comorbidity exactly. Moreover, Selective serotonin reuptake inhibitors, insulin and metformin, which are recommended for treating diabetes mellitus-induced depression, were found to be a risk factor in some complications of diabetes. AIMS Given these problems, many researchers made remarkable efforts to analyze diabetes complicating depression from different aspects, including insulin resistance, stress and Hypothalamic-Pituitary-Adrenal axis, neurological system, oxidative stress, and inflammation. Drug therapy, such as Hydrogen Sulfide, Cannabidiol, Ascorbic Acid and Hesperidin, are conducive to alleviating diabetes mellitus and depression. Here, we reviewed the exact pathophysiology underlying the comorbidity between depressive disorder and diabetes mellitus and drug therapy. METHODS The review refers to the available literature in PubMed and Web of Science, searching critical terms related to diabetes mellitus, depression and drug therapy. RESULTS In this review, we found that brain structure and function, neurogenesis, brain-derived neurotrophic factor and glucose and lipid metabolism were involved in the pathophysiology of the comorbidity. Obesity might lead to diabetes mellitus and depression through reduced adiponectin and increased leptin and resistin. In addition, drug therapy displayed in this review could expand the region of potential therapy. CONCLUSIONS The review summarizes the mechanisms underlying the comorbidity. It also overviews drug therapy with anti-diabetic and anti-depressant effects.
Collapse
Affiliation(s)
- Sixin Li
- Department of Psychiatry, The School of Clinical MedicineHunan University of Chinese MedicineChangshaHunanChina
- Department of PsychiatryBrain Hospital of Hunan Province (The Second People's Hospital of Hunan Province)ChangshaHunanChina
| | - Dong Yang
- Department of Psychiatry, The School of Clinical MedicineHunan University of Chinese MedicineChangshaHunanChina
- Department of PsychiatryBrain Hospital of Hunan Province (The Second People's Hospital of Hunan Province)ChangshaHunanChina
| | - Xuhui Zhou
- Department of Psychiatry, The School of Clinical MedicineHunan University of Chinese MedicineChangshaHunanChina
- Department of PsychiatryBrain Hospital of Hunan Province (The Second People's Hospital of Hunan Province)ChangshaHunanChina
| | - Lu Chen
- Department of Gastroenterology, The School of Clinical MedicineHunan University of Chinese MedicineChangshaHunanChina
- Department of GastroenterologyBrain Hospital of Hunan Province (The Second People's Hospital of Hunan Province)ChangshaHunanChina
| | - Lini Liu
- Department of Psychiatry, The School of Clinical MedicineHunan University of Chinese MedicineChangshaHunanChina
- Department of PsychiatryBrain Hospital of Hunan Province (The Second People's Hospital of Hunan Province)ChangshaHunanChina
| | - Ruoheng Lin
- Department of Psychiatry, National Clinical Research Center for Mental DisordersThe Second Xiangya Hospital of Central South UniversityChangshaHunanChina
| | - Xinyu Li
- Department of Psychiatry, The School of Clinical MedicineHunan University of Chinese MedicineChangshaHunanChina
- Department of PsychiatryBrain Hospital of Hunan Province (The Second People's Hospital of Hunan Province)ChangshaHunanChina
| | - Ying Liu
- Department of Psychiatry, The School of Clinical MedicineHunan University of Chinese MedicineChangshaHunanChina
- Department of PsychiatryBrain Hospital of Hunan Province (The Second People's Hospital of Hunan Province)ChangshaHunanChina
| | - Huiwen Qiu
- Department of Psychiatry, The School of Clinical MedicineHunan University of Chinese MedicineChangshaHunanChina
- Department of PsychiatryBrain Hospital of Hunan Province (The Second People's Hospital of Hunan Province)ChangshaHunanChina
| | - Hui Cao
- Department of Psychiatry, The School of Clinical MedicineHunan University of Chinese MedicineChangshaHunanChina
- Department of PsychiatryBrain Hospital of Hunan Province (The Second People's Hospital of Hunan Province)ChangshaHunanChina
| | - Jian Liu
- Center for Medical Research and Innovation, The First Hospital, Hunan University of Chinese MedicineChangshaHunanChina
| | - Quan Cheng
- Department of Neurosurgery, Xiangya HospitalCentral South UniversityChangshaHunanChina
- National Clinical Research Center for Geriatric Disorders, Xiangya HospitalCentral South UniversityChangshaHunanChina
| |
Collapse
|
22
|
Li J, Zheng G, Jiang D, Deng C, Zhang Y, Ma Y, Su J. Mendelian randomization analysis reveals a causal effect of Streptococcus salivarius on diabetic retinopathy through regulating host fasting glucose. J Cell Mol Med 2024; 28:e18200. [PMID: 38506069 PMCID: PMC10951888 DOI: 10.1111/jcmm.18200] [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: 11/30/2023] [Revised: 01/29/2024] [Accepted: 02/20/2024] [Indexed: 03/21/2024] Open
Abstract
Diabetic retinopathy (DR) is one of leading causes of vision loss in adults with increasing prevalence worldwide. Increasing evidence has emphasized the importance of gut microbiome in the aetiology and development of DR. However, the causal relationship between gut microbes and DR remains largely unknown. To investigate the causal associations of DR with gut microbes and DR risk factors, we employed two-sample Mendelian Randomization (MR) analyses to estimate the causal effects of 207 gut microbes on DR outcomes. Inputs for MR included Genome-wide Association Study (GWAS) summary statistics of 207 taxa of gut microbes (the Dutch Microbiome Project) and 21 risk factors for DR. The GWAS summary statistics data of DR was from the FinnGen Research Project. Data analysis was performed in May 2023. We identified eight bacterial taxa that exhibited significant causal associations with DR (FDR < 0.05). Among them, genus Collinsella and species Collinsella aerofaciens were associated with increased risk of DR, while the species Bacteroides faecis, Burkholderiales bacterium_1_1_47, Ruminococcus torques, Streptococcus salivarius, genus Burkholderiales_noname and family Burkholderiales_noname showed protective effects against DR. Notably, we found that the causal effect of species Streptococcus salivarius on DR was mediated through the level of host fasting glucose, a well-established risk factor for DR. Our results reveal that specific gut microbes may be causally linked to DR via mediating host metabolic risk factors, highlighting potential novel therapeutic or preventive targets for DR.
Collapse
Affiliation(s)
- Jingjing Li
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye HospitalWenzhou Medical UniversityWenzhouChina
- Department of Biomedical Informatics, Institute of Biomedical Big DataWenzhou Medical UniversityWenzhouChina
| | - Gongwei Zheng
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye HospitalWenzhou Medical UniversityWenzhouChina
- Department of Biomedical Informatics, Institute of Biomedical Big DataWenzhou Medical UniversityWenzhouChina
| | - Dingping Jiang
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye HospitalWenzhou Medical UniversityWenzhouChina
- Department of Biomedical Informatics, Institute of Biomedical Big DataWenzhou Medical UniversityWenzhouChina
| | - Chunyu Deng
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye HospitalWenzhou Medical UniversityWenzhouChina
- Department of Biomedical Informatics, Institute of Biomedical Big DataWenzhou Medical UniversityWenzhouChina
| | - Yaru Zhang
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye HospitalWenzhou Medical UniversityWenzhouChina
- Department of Biomedical Informatics, Institute of Biomedical Big DataWenzhou Medical UniversityWenzhouChina
| | - Yunlong Ma
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye HospitalWenzhou Medical UniversityWenzhouChina
- Department of Biomedical Informatics, Institute of Biomedical Big DataWenzhou Medical UniversityWenzhouChina
| | - Jianzhong Su
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye HospitalWenzhou Medical UniversityWenzhouChina
- Department of Biomedical Informatics, Institute of Biomedical Big DataWenzhou Medical UniversityWenzhouChina
- Oujiang LaboratoryZhejiang Lab for Regenerative Medicine, Vision and Brain HealthWenzhouChina
- Wenzhou InstituteUniversity of Chinese Academy of SciencesWenzhouChina
| |
Collapse
|
23
|
Zhou Z, Yao Y, Sun Y, Wang X, Huang S, Hou J, Wang L, Wei F. Serum betaine and dimethylglycine in mid-pregnancy and the risk of gestational diabetes mellitus: a case-control study. Endocrine 2024:10.1007/s12020-024-03732-4. [PMID: 38448678 DOI: 10.1007/s12020-024-03732-4] [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: 10/02/2023] [Accepted: 02/04/2024] [Indexed: 03/08/2024]
Abstract
PURPOSE To investigate the associations of choline, betaine, dimethylglycine (DMG), L-carnitine, and Trimethylamine-N-oxide (TMAO) with the risk of Gestational diabetes mellitus (GDM) as well as the markers of glucose homeostasis. METHODS We performed a case-control study including 200 diagnosed GDM cases and 200 controls matched by maternal age (±2 years) and gestational age (±2 weeks). Concentrations of serum metabolites were measured by the high-performance liquid chromatography - tandem mass spectrometry (HPLC-MS/MS). RESULTS Compared to the control group, GDM group had significantly lower serum betaine concentration and betaine/choline ratio, and higher DMG concentration. Furthermore, decreased betaine concentration and betaine/choline ratio, increased DMG concentration showed significant association with the risk of GDM. In addition, serum betaine concentrations were negatively associated with blood glucose levels at 1-h post-glucose load (OGTT-1h), and both betaine and L-carnitine concentrations were positively associated with 1,5-anhydroglucitol levels. Betaine/choline ratio was negatively associated with OGTT-1h and blood glucose levels at 2-h post-glucose load (OGTT-2h) and serum choline concentrations were negatively associated with fasting blood glucose and positively associated with OGTT-2h. CONCLUSION Decreased serum betaine concentrations and betaine/choline ratio, and elevated DMG concentrations could be significant risk factors for GDM. Furthermore, betaine may be associated with blood glucose regulation and short-term glycemic fluctuations.
Collapse
Affiliation(s)
- Ziqing Zhou
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, Anhui Province, China
- The Genetics Laboratory, Longgang District Maternity & Child Healthcare Hospital of Shenzhen City (Longgang Maternity and Child Institute of Shantou University Medical College), Shenzhen, Guangdong Province, China
| | - Yao Yao
- The Genetics Laboratory, Longgang District Maternity & Child Healthcare Hospital of Shenzhen City (Longgang Maternity and Child Institute of Shantou University Medical College), Shenzhen, Guangdong Province, China
| | - Yanan Sun
- The Genetics Laboratory, Longgang District Maternity & Child Healthcare Hospital of Shenzhen City (Longgang Maternity and Child Institute of Shantou University Medical College), Shenzhen, Guangdong Province, China
- Medical Insurance Office of Shenzhen Longgang Central Hospital, Shenzhen, Guangdong Province, China
| | - Xin Wang
- The Genetics Laboratory, Longgang District Maternity & Child Healthcare Hospital of Shenzhen City (Longgang Maternity and Child Institute of Shantou University Medical College), Shenzhen, Guangdong Province, China
- Jiamusi University, Jiamusi, Heilongjiang Province, China
| | - Shang Huang
- The Genetics Laboratory, Longgang District Maternity & Child Healthcare Hospital of Shenzhen City (Longgang Maternity and Child Institute of Shantou University Medical College), Shenzhen, Guangdong Province, China
- Shenzhen Children's Hospital of China Medical University, Shenzhen, Guangdong Province, China
| | - Jianli Hou
- Department of Gynecology and Obstetrics, Longgang District Maternity & Child Healthcare Hospital of Shenzhen City (Longgang Maternity and Child Institute of Shantou University Medical College), Shenzhen, Guangdong Province, China
| | - Lijun Wang
- Department of Nutrition, School of Medicine, Jinan University, Guangzhou, Guangdong Province, China.
| | - Fengxiang Wei
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, Anhui Province, China.
- The Genetics Laboratory, Longgang District Maternity & Child Healthcare Hospital of Shenzhen City (Longgang Maternity and Child Institute of Shantou University Medical College), Shenzhen, Guangdong Province, China.
| |
Collapse
|
24
|
Sastre M, Cimbalo A, Mañes J, Manyes L. Gut Microbiota and Nutrition: Strategies for the Prevention and Treatment of Type 2 Diabetes. J Med Food 2024; 27:97-109. [PMID: 38381517 DOI: 10.1089/jmf.2022.0154] [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: 02/22/2024] Open
Abstract
The prevalence of diabetes has increased in last decades worldwide and is expected to continue to do so in the coming years, reaching alarming figures. Evidence have shown that patients with type 2 diabetes (T2D) have intestinal microbial dysbiosis. Moreover, several mechanisms link the microbiota with the appearance of insulin resistance and diabetes. Diet is a crucial factor related to changes in the composition, diversity, and activity of gut microbiota (GM). In this review, the current and future possibilities of nutrient-GM interactions as a strategy to alleviate T2D are discussed, as well as the mechanisms related to decreased low-grade inflammation and insulin resistance. A bibliographic search of clinical trials in Pubmed, Web of Science, and Scopus was carried out, using the terms "gut microbiota, diet and diabetes." The data analyzed in this review support the idea that dietary interventions targeting changes in the microbiota, including the use of prebiotics and probiotics, can improve glycemic parameters. However, these strategies should be individualized taking into account other internal and external factors. Advances in the understanding of the role of the microbiota in the development of metabolic diseases such as T2D, and its translation into a therapeutic approach for the management of diabetes, are necessary to allow a comprehensive approach.
Collapse
Affiliation(s)
- Maria Sastre
- Laboratory of Food Chemistry and Toxicology, University of Valencia, Valencia, Spain
| | - Alessandra Cimbalo
- Laboratory of Food Chemistry and Toxicology, University of Valencia, Valencia, Spain
| | - Jordi Mañes
- Laboratory of Food Chemistry and Toxicology, University of Valencia, Valencia, Spain
| | - Lara Manyes
- Laboratory of Food Chemistry and Toxicology, University of Valencia, Valencia, Spain
| |
Collapse
|
25
|
Niu C, Lv W, Zhu X, Dong Z, Yuan K, Jin Q, Zhang P, Li P, Mao M, Dong T, Chen Z, Luo J, Hou L, Zhang C, Hao K, Chen S, Huang Z. Intestinal Translocation of Live Porphyromonas gingivalis Drives Insulin Resistance. J Dent Res 2024; 103:197-207. [PMID: 38185909 DOI: 10.1177/00220345231214195] [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: 01/09/2024] Open
Abstract
Periodontitis has been emphasized as a risk factor of insulin resistance-related systemic diseases. Accumulating evidence has suggested a possible "oral-gut axis" linking oral infection and extraoral diseases, but it remains unclear whether periodontal pathogens can survive the barriers of the digestive tract and how they play their pathogenic roles. The present study established a periodontitis mouse model through oral ligature plus Porphyromonas gingivalis inoculation and demonstrated that periodontitis aggravated diet-induced obesity and insulin resistance, while also causing P. gingivalis enrichment in the intestine. Metabolic labeling strategy validated that P. gingivalis could translocate to the gastrointestinal tract in a viable state. Oral administration of living P. gingivalis elicited insulin resistance, while administration of pasteurized P. gingivalis had no such effect. Combination analysis of metagenome sequencing and nontargeted metabolomics suggested that the tryptophan metabolism pathway, specifically indole and its derivatives, was involved in the pathogenesis of insulin resistance caused by oral administration of living P. gingivalis. Moreover, liquid chromatography-high-resolution mass spectrometry analysis confirmed that the aryl hydrocarbon receptor (AhR) ligands, mainly indole acetic acid, tryptamine, and indole-3-aldehyde, were reduced in diet-induced obese mice with periodontitis, leading to inactivation of AhR signaling. Supplementation with Ficz (6-formylindolo (3,2-b) carbazole), an AhR agonist, alleviated periodontitis-associated insulin resistance, in which the restoration of gut barrier function might play an important role. Collectively, these findings reveal that the oral-gut translocation of viable P. gingivalis works as a fuel linking periodontitis and insulin resistance, in which reduction of AhR ligands and inactivation of AhR signaling are involved. This study provides novel insight into the role of the oral-gut axis in the pathogenesis of periodontitis-associated comorbidities.
Collapse
Affiliation(s)
- C Niu
- Department of Endodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P. R. China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, P. R. China
- National Clinical Research Center for Oral Diseases, National Center for Stomatology, Shanghai, P. R. China
- Shanghai Key Laboratory of Stomatology, Shanghai, P. R. China
| | - W Lv
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, P. R. China
| | - X Zhu
- Department of Endodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P. R. China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, P. R. China
- National Clinical Research Center for Oral Diseases, National Center for Stomatology, Shanghai, P. R. China
- Shanghai Key Laboratory of Stomatology, Shanghai, P. R. China
| | - Z Dong
- Department of Oral Implantology, Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai, P. R. China
| | - K Yuan
- Department of Endodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P. R. China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, P. R. China
- National Clinical Research Center for Oral Diseases, National Center for Stomatology, Shanghai, P. R. China
- Shanghai Key Laboratory of Stomatology, Shanghai, P. R. China
| | - Q Jin
- Department of Endodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P. R. China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, P. R. China
- National Clinical Research Center for Oral Diseases, National Center for Stomatology, Shanghai, P. R. China
- Shanghai Key Laboratory of Stomatology, Shanghai, P. R. China
| | - P Zhang
- Department of Endodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P. R. China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, P. R. China
- National Clinical Research Center for Oral Diseases, National Center for Stomatology, Shanghai, P. R. China
- Shanghai Key Laboratory of Stomatology, Shanghai, P. R. China
| | - P Li
- Department of Endodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P. R. China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, P. R. China
- National Clinical Research Center for Oral Diseases, National Center for Stomatology, Shanghai, P. R. China
- Shanghai Key Laboratory of Stomatology, Shanghai, P. R. China
| | - M Mao
- Department of Endodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P. R. China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, P. R. China
- National Clinical Research Center for Oral Diseases, National Center for Stomatology, Shanghai, P. R. China
- Shanghai Key Laboratory of Stomatology, Shanghai, P. R. China
| | - T Dong
- Department of Endodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P. R. China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, P. R. China
- National Clinical Research Center for Oral Diseases, National Center for Stomatology, Shanghai, P. R. China
- Shanghai Key Laboratory of Stomatology, Shanghai, P. R. China
| | - Z Chen
- Department of Endodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P. R. China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, P. R. China
- National Clinical Research Center for Oral Diseases, National Center for Stomatology, Shanghai, P. R. China
- Shanghai Key Laboratory of Stomatology, Shanghai, P. R. China
| | - J Luo
- Department of Endodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P. R. China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, P. R. China
- National Clinical Research Center for Oral Diseases, National Center for Stomatology, Shanghai, P. R. China
- Shanghai Key Laboratory of Stomatology, Shanghai, P. R. China
| | - L Hou
- Department of Nursing, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, P. R. China
| | - C Zhang
- Department of Oral Implantology, Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai, P. R. China
| | - K Hao
- Department of Oral Implantology, Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai, P. R. China
| | - S Chen
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, P. R. China
- Department of Oral Implantology, Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai, P. R. China
| | - Z Huang
- Department of Endodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P. R. China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, P. R. China
- National Clinical Research Center for Oral Diseases, National Center for Stomatology, Shanghai, P. R. China
- Shanghai Key Laboratory of Stomatology, Shanghai, P. R. China
| |
Collapse
|
26
|
Jiang X, Zhang B, Lan F, Zhong C, Jin J, Li X, Zhou Q, Li J, Yang N, Wen C, Sun C. Host genetics and gut microbiota jointly regulate blood biochemical indicators in chickens. Appl Microbiol Biotechnol 2023; 107:7601-7620. [PMID: 37792060 PMCID: PMC10656342 DOI: 10.1007/s00253-023-12814-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 09/14/2023] [Accepted: 09/22/2023] [Indexed: 10/05/2023]
Abstract
Blood biochemical indicators play a crucial role in assessing an individual's overall health status and metabolic function. In this study, we measured five blood biochemical indicators, including total cholesterol (CHOL), low-density lipoprotein cholesterol (LDL-CH), triglycerides (TG), high-density lipoprotein cholesterol (HDL-CH), and blood glucose (BG), as well as 19 growth traits of 206 male chickens. By integrating host whole-genome information and 16S rRNA sequencing of the duodenum, jejunum, ileum, cecum, and feces microbiota, we assessed the contributions of host genetics and gut microbiota to blood biochemical indicators and their interrelationships. Our results demonstrated significant negative phenotypic and genetic correlations (r = - 0.20 ~ - 0.67) between CHOL and LDL-CH with growth traits such as body weight, abdominal fat content, muscle content, and shin circumference. The results of heritability and microbiability indicated that blood biochemical indicators were jointly regulated by host genetics and gut microbiota. Notably, the heritability of HDL-CH was estimated to be 0.24, while the jejunal microbiability for BG and TG reached 0.45 and 0.23. Furthermore, by conducting genome-wide association study (GWAS) with the single-nucleotide polymorphism (SNPs), insertion/deletion (indels), and structural variation (SV), we identified RAP2C, member of the RAS oncogene family (RAP2C), dedicator of cytokinesis 11 (DOCK11), neurotensin (NTS) and BOP1 ribosomal biogenesis factor (BOP1) as regulators of HDL-CH, and glycerophosphodiester phosphodiesterase domain containing 5 (GDPD5), dihydrodiol dehydrogenase (DHDH), and potassium voltage-gated channel interacting protein 1 (KCNIP1) as candidate genes of BG. Moreover, our findings suggest that cecal RF39 and Clostridia_UCG_014 may be linked to the regulation of CHOL, and jejunal Streptococcaceae may be involved in the regulation of TG. Additionally, microbial GWAS results indicated that the presence of gut microbiota was under host genetic regulation. Our findings provide valuable insights into the complex interaction between host genetics and microbiota in shaping the blood biochemical profile of chickens. KEY POINTS: • Multiple candidate genes were identified for the regulation of CHOL, HDL-CH, and BG. • RF39, Clostridia_UCG_014, and Streptococcaceae were implicated in CHOL and TG modulation. • The composition of gut microbiota is influenced by host genetics.
Collapse
Affiliation(s)
- Xinwei Jiang
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Boxuan Zhang
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Fangren Lan
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Conghao Zhong
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Jiaming Jin
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Xiaochang Li
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Qianqian Zhou
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Junying Li
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Ning Yang
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Chaoliang Wen
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China.
| | - Congjiao Sun
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
27
|
Wang A, Guan B, Zhang H, Xu H. Danger-associated metabolites trigger metaflammation: A crowbar in cardiometabolic diseases. Pharmacol Res 2023; 198:106983. [PMID: 37931790 DOI: 10.1016/j.phrs.2023.106983] [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: 08/01/2023] [Revised: 10/12/2023] [Accepted: 11/03/2023] [Indexed: 11/08/2023]
Abstract
Cardiometabolic diseases (CMDs) are characterized by a series of metabolic disorders and chronic low-grade inflammation. CMDs contribute to a high burden of mortality and morbidity worldwide. Host-microbial metabolic regulation that triggers metaflammation is an emerging field of study that promotes a new perspective for perceiving cardiovascular risks. The term metaflammation denotes the entire cascade of immune responses activated by a new class of metabolites known as "danger-associated metabolites" (DAMs). It is being proposed by the present review for the first time. We summarize current studies covering bench to bedside aspects of DAMs to better understand CMDs in the context of DAMs. We have focused on the involvement of DAMs in the pathophysiological development of CMDs, including the disruption of immune homeostasis and chronic inflammation-triggered damage leading to CMD-related adverse events, as well as emerging therapeutic approaches for targeting DAM metabolism in CMDs.
Collapse
Affiliation(s)
- Anlu Wang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China; National Clinical Research Center for Chinese Medicine Cardiology, Beijing 100091, China
| | - Baoyi Guan
- Department of Internal Medicine-Cardiovascular, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510000, China
| | - He Zhang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China; National Clinical Research Center for Chinese Medicine Cardiology, Beijing 100091, China
| | - Hao Xu
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China; National Clinical Research Center for Chinese Medicine Cardiology, Beijing 100091, China.
| |
Collapse
|
28
|
Lv Q, Zhou J, Wang C, Yang X, Han Y, Zhou Q, Yao R, Sui A. A dynamics association study of gut barrier and microbiota in hyperuricemia. Front Microbiol 2023; 14:1287468. [PMID: 38088975 PMCID: PMC10711221 DOI: 10.3389/fmicb.2023.1287468] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 11/09/2023] [Indexed: 02/28/2024] Open
Abstract
Introduction The intricate interplay between gut microbiota and hyperuricemia remains a subject of growing interest. However, existing studies only provided snapshots of the gut microbiome at single time points, the temporal dynamics of gut microbiota alterations during hyperuricemia progression and the intricate interplay between the gut barrier and microbiota remain underexplored. Our investigation revealed compelling insights into the dynamic changes in both gut microbiota and intestinal barrier function throughout the course of hyperuricemia. Methods The hyperuricemia mice (HY) were given intragastric administration of adenine and potassium oxalate. Gut microbiota was analyzed by 16S rRNA sequencing at 3, 7, 14, and 21 days after the start of the modeling process. Intestinal permeability as well as LPS, TNF-α, and IL-1β levels were measured at 3, 7, 14, and 21 days. Results We discovered that shifts in microbial community composition occur prior to the onset of hyperuricemia, key bacterial Bacteroidaceae, Bacteroides, and Blautia exhibited reduced levels, potentially fueling microbial dysbiosis as the disease progresses. During the course of hyperuricemia, the dynamic fluctuations in both uric acid levels and intestinal barrier function was accompanied with the depletion of key beneficial bacteria, including Prevotellaceae, Muribaculum, Parabacteroides, Akkermansia, and Bacteroides, and coincided with an increase in pathogenic bacteria such as Oscillibacter and Ruminiclostridium. This microbial community shift likely contributed to elevated lipopolysaccharide (LPS) and pro-inflammatory cytokine levels, ultimately promoting metabolic inflammation. The decline of Burkholderiaceae and Parasutterella was inversely related to uric acid levels, Conversely, key families Ruminococcaceae, Family_XIII, genera Anaeroplasma exhibited positive correlations with uric acid levels. Akkermansiaceae and Bacteroidaceae demonstrating negative correlations, while LPS-containing microbiota such as Desulfovibrio and Enterorhabdus exhibited positive correlations with intestinal permeability. Conclusion In summary, this study offers a dynamic perspective on the complex interplay between gut microbiota, uric acid levels, and intestinal barrier function during hyperuricemia progression. Our study suggested that Ruminiclostridium, Bacteroides, Akkermansiaceae, Bilophila, Burkholderiaceae and Parasutterella were the key bacteria that play vital rols in the progress of hyperuricemia and compromised intestinal barrier, which provide a potential avenue for therapeutic interventions in hyperuricemia.
Collapse
Affiliation(s)
- Qiulan Lv
- Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jun Zhou
- Laboratory Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Changyao Wang
- Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiaomin Yang
- Laboratory Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yafei Han
- Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Quan Zhou
- Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ruyong Yao
- Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Aihua Sui
- Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
29
|
Zhou H, Luo Y, Zhang W, Xie F, Deng C, Zheng W, Zhu S, Wang Q. Causal effect of gut-microbiota-derived metabolite trimethylamine N-oxide on Parkinson's disease: A Mendelian randomization study. Eur J Neurol 2023; 30:3451-3461. [PMID: 36692876 DOI: 10.1111/ene.15702] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 01/01/2023] [Accepted: 01/19/2023] [Indexed: 01/25/2023]
Abstract
BACKGROUND AND PURPOSE It has been suggested that trimethylamine N-oxide (TMAO) is related to Parkinson's disease (PD) in observational studies. However, the direction of this association is inconsistent. An exploratory Mendelian randomization study was conducted to investigate whether TMAO and its precursors have a causal relationship with PD. METHODS Summary statistics were obtained for single nucleotide polymorphisms related to circulating levels of TMAO, betaine, carnitine and choline, and the corresponding data for the risk, age at onset and progression of PD from genome-wide association studies. Inverse-variance weighting was used as the primary method for effect estimation. The false discovery rate was applied to the correction of multiple testing. A p value of association <0.05 but above the false discovery rate corrected threshold was deemed suggestive evidence of a possible association. A range of robust Mendelian randomization methods were used for sensitivity analysis. RESULTS Suggestive evidence was observed of an inverse causal effect of TMAO on motor fluctuations (odds ratio [OR] 0.851, 95% confidence interval [CI] 0.731, 0.990, p = 0.037) and carnitine on insomnia (OR 0.817, 95% CI 0.700, 0.954, p = 0.010) and a positive causal effect of betaine on Hoehn-Yahr stage (OR 1.397, 95% CI 1.112, 1.756, p = 0.004), Unified Parkinson's Disease Rating Scale (UPDRS) III score (β = 0.138, 95% CI 0.051, 0.225, p = 0.002), motor fluctuations (OR 1.236, 95% CI 1.011, 1.511, p = 0.039), and choline on UPDRS IV (β = 0.106, 95% CI 0.026, 0.185, p = 0.009) and modified Schwab and England Activities of Daily Living Scale score (β = 0.806, 95% CI 0.127, 1.484, p = 0.020). CONCLUSIONS Our findings provide suggestive evidence that TMAO and its precursors have a causal effect on the progression of PD. Further investigation of the underlying mechanisms is required.
Collapse
Affiliation(s)
- Hang Zhou
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Guangzhou, China
| | - Yuqi Luo
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Guangzhou, China
| | - Wenjie Zhang
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Guangzhou, China
| | - Fen Xie
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Guangzhou, China
| | - Chao Deng
- School of Medical, Indigenous and Health Sciences, and Molecular Horizons, University of Wollongong, Wollongong, Australia
| | - Wenhua Zheng
- Centre of Reproduction, Development and Aging and Institute of Translation Medicine, Faculty of Health Sciences, University of Macau, Taipa, China
| | - Shuzhen Zhu
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Guangzhou, China
| | - Qing Wang
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Guangzhou, China
| |
Collapse
|
30
|
Zhang Y, Wang J, Ge W, Song Y, He R, Wang Z, Zhao L. Camel milk peptides alleviate hyperglycemia by regulating gut microbiota and metabolites in type 2 diabetic mice. Food Res Int 2023; 173:113278. [PMID: 37803591 DOI: 10.1016/j.foodres.2023.113278] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 07/11/2023] [Accepted: 07/12/2023] [Indexed: 10/08/2023]
Abstract
This study aimed to investigate the hypoglycemic effect of Camel milk peptides (CMPs) on Type 2 diabetes mellitus (T2DM) mice and reveal its related mechanism from the aspect of gut microbiota and metabolites. The administering CMPs significantly alleviated the weight loss, polydipsia and polyphagia, reduced fasting blood glucose (FBG), improved insulin resistance and sensitivity, and restored the level of serum hormones, lipopolysaccharide (LPS), lipid metabolic and tissue damage. Furthermore, CMPs intervention remarkably reversed gut microbiota dysbiosis in T2DM mice by reducing the relative abundance of Proteobacteria, Allobaculum, Clostridium, Shigella and the Firmicutes/Bacteroidetes ratio, while increasing the relative abundance of Bacteroidetes and Blautia. Metabolomic analysis identified 84 different metabolites between T2DM and CMPs-treated groups, participating in three pathways of Pantothenate and CoA biosynthesis, Phenylalanine metabolism and Linoleic acid metabolism. Ureidopropionic acid, pantothenic acid, hippuric acid, hydrocinnamic acid and linoleic acid were identified as key acidic metabolites closely related to hypoglycemic effect. Correlation analysis indicated that CMPs might have a hypoglycemic effect through their impact on gut microbiota, leading to variations in short-chain fatty acids (SCFAs), acidic metabolites and metabolic pathways. These findings suggest that CMPs could be a beneficial nutritional supplement for intervention T2DM.
Collapse
Affiliation(s)
- Yongjin Zhang
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Ju Wang
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Wupeng Ge
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Yuxuan Song
- Shaanxi Engineering Research Centre of Dairy Products Quality, Safety and Health, Northwest A&F University, Yangling 712100, China
| | - Rui He
- Shaanxi Baiyue Youlishi Dairy Industry Co. Ltd., Xianyang 712000, China
| | - Zhi Wang
- Shaanxi Baiyue Youlishi Dairy Industry Co. Ltd., Xianyang 712000, China
| | - Lili Zhao
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi 712100, China.
| |
Collapse
|
31
|
Liu P, Li H, Xu H, Gong J, Jiang M, Xu Z, Shi J. Aggravated hepatic fibrosis induced by phenylalanine and tyrosine was ameliorated by chitooligosaccharides supplementation. iScience 2023; 26:107754. [PMID: 37731617 PMCID: PMC10507131 DOI: 10.1016/j.isci.2023.107754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 03/21/2023] [Accepted: 08/24/2023] [Indexed: 09/22/2023] Open
Abstract
Hepatic fibrosis is a classic pathological manifestation of metabolic chronic hepatopathy. The pathological process might either gradually deteriorate into cirrhosis and ultimately liver cancer with inappropriate nutrition supply, or be slowed down by several multifunctional nutrients, alternatively. Herein, we found diet with excessive phenylalanine (Phe) and tyrosine (Tyr) exacerbated hepatic fibrosis symptoms of liver dysfunction and gut microflora dysbiosis in mice. Chitooligosaccharides (COS) could ameliorate hepatic fibrosis with the regulation of amino acid metabolism by downregulating the mTORC1 pathway, especially that of Phe and Tyr, and also with the alleviation of the dysbiosis of gut microbiota, simultaneously. Conclusively, this work presents new insight into the role of Phe and Tyr in the pathologic process of hepatic fibrosis, while revealing the effectiveness and molecular mechanism of COS in improving hepatic fibrosis from the perspective of metabolites.
Collapse
Affiliation(s)
- Peng Liu
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, China
- Institute of Edible Fungi, Shanghai Academy of Agricultural Sciences, 1000 Jinqi Road, Shanghai 201403, China
| | - Heng Li
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, China
| | - Hongyu Xu
- National Engineering Research Center for Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi 214122, China
| | - Jinsong Gong
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, China
| | - Min Jiang
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, China
| | - Zhenghong Xu
- National Engineering Research Center for Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi 214122, China
| | - Jinsong Shi
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, China
| |
Collapse
|
32
|
Mihuta MS, Paul C, Borlea A, Roi CM, Pescari D, Velea-Barta OA, Mozos I, Stoian D. Connections between serum Trimethylamine N-Oxide (TMAO), a gut-derived metabolite, and vascular biomarkers evaluating arterial stiffness and subclinical atherosclerosis in children with obesity. Front Endocrinol (Lausanne) 2023; 14:1253584. [PMID: 37850094 PMCID: PMC10577381 DOI: 10.3389/fendo.2023.1253584] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 09/12/2023] [Indexed: 10/19/2023] Open
Abstract
Introduction Childhood obesity leads to early subclinical atherosclerosis and arterial stiffness. Studying biomarkers like trimethylamine N-oxide (TMAO), linked to cardio-metabolic disorders in adults, is crucial to prevent long-term cardiovascular issues. Methods The study involved 70 children aged 4 to 18 (50 obese, 20 normal-weight). Clinical examination included BMI, waist measurements, puberty stage, the presence of acanthosis nigricans, and irregular menstrual cycles. Subclinical atherosclerosis was assessed by measuring the carotid intima-media thickness (CIMT), and the arterial stiffness was evaluated through surrogate markers like the pulse wave velocity (PWV), augmentation index (AIx), and peripheral and central blood pressures. The blood biomarkers included determining the values of TMAO, HOMA-IR, and other usual biomarkers investigating metabolism. Results The study detected significantly elevated levels of TMAO in obese children compared to controls. TMAO presented positive correlations to BMI, waist circumference and waist-to-height ratio and was also observed as an independent predictor of all three parameters. Significant correlations were observed between TMAO and vascular markers such as CIMT, PWV, and peripheral BP levels. TMAO independently predicts CIMT, PWV, peripheral BP, and central SBP levels, even after adding BMI, waist circumference, waist-to-height ratio, puberty development and age in the regression model. Obese children with high HOMA-IR presented a greater weight excess and significantly higher vascular markers, but TMAO levels did not differ significantly from the obese with HOMA-IR Conclusion Our study provides compelling evidence supporting the link between serum TMAO, obesity, and vascular damage in children. These findings highlight the importance of further research to unravel the underlying mechanisms of this connection.
Collapse
Affiliation(s)
- Monica Simina Mihuta
- Department of Doctoral Studies, Victor Babes University of Medicine and Pharmacy, Timisoara, Romania
- Center of Molecular Research in Nephrology and Vascular Disease, Faculty of Medicine, Victor Babes University of Medicine and Pharmacy, Timisoara, Romania
| | - Corina Paul
- Department of Pediatrics, Victor Babes University of Medicine and Pharmacy, Timisoara, Romania
| | - Andreea Borlea
- Center of Molecular Research in Nephrology and Vascular Disease, Faculty of Medicine, Victor Babes University of Medicine and Pharmacy, Timisoara, Romania
- 2nd Department of Internal Medicine, Victor Babes University of Medicine and Pharmacy, Timisoara, Romania
| | - Cristina Mihaela Roi
- Department of Doctoral Studies, Victor Babes University of Medicine and Pharmacy, Timisoara, Romania
- Center of Molecular Research in Nephrology and Vascular Disease, Faculty of Medicine, Victor Babes University of Medicine and Pharmacy, Timisoara, Romania
| | - Denisa Pescari
- Department of Doctoral Studies, Victor Babes University of Medicine and Pharmacy, Timisoara, Romania
| | - Oana-Alexandra Velea-Barta
- 3rd Department of Odontotherapy and Endodontics, Faculty of Dental Medicine, Victor Babes University of Medicine and Pharmacy, Timisoara, Romania
| | - Ioana Mozos
- Department of Functional Sciences—Pathophysiology, Center for Translational Research and Systems Medicine, Victor Babes University of Medicine and Pharmacy, Timisoara, Romania
| | - Dana Stoian
- Center of Molecular Research in Nephrology and Vascular Disease, Faculty of Medicine, Victor Babes University of Medicine and Pharmacy, Timisoara, Romania
- 2nd Department of Internal Medicine, Victor Babes University of Medicine and Pharmacy, Timisoara, Romania
| |
Collapse
|
33
|
Wang R, Wang QY, Bai Y, Bi YG, Cai SJ. Research progress of diabetic retinopathy and gut microecology. Front Microbiol 2023; 14:1256878. [PMID: 37744925 PMCID: PMC10513461 DOI: 10.3389/fmicb.2023.1256878] [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: 07/12/2023] [Accepted: 08/23/2023] [Indexed: 09/26/2023] Open
Abstract
According to the prediction of the International Diabetes Federation, global diabetes mellitus (DM) patients will reach 783.2 million in 2045. The increasing incidence of DM has led to a global epidemic of diabetic retinopathy (DR). DR is a common microvascular complication of DM, which has a significant impact on the vision of working-age people and is one of the main causes of blindness worldwide. Substantial research has highlighted that microangiopathy and chronic low-grade inflammation are widespread in the retina of DR. Meanwhile, with the introduction of the gut-retina axis, it has also been found that DR is associated with gut microecological disorders. The disordered structure of the GM and the destruction of the gut barrier result in the release of abnormal GM flora metabolites into the blood circulation. In addition, this process induced alterations in the expression of various cytokines and proteins, which further modulate the inflammatory microenvironment, vascular damage, oxidative stress, and immune levels within the retina. Such alterations led to the development of DR. In this review, we discuss the corresponding alterations in the structure of the GM flora and its metabolites in DR, with a more detailed focus on the mechanism of gut microecology in DR. Finally, we summarize the potential therapeutic approaches of DM/DR, mainly regulating the disturbed gut microecology to restore the homeostatic level, to provide a new perspective on the prevention, monitoring, and treatment of DR.
Collapse
Affiliation(s)
- Rui Wang
- Department of Ophthalmology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Special Key Laboratory of Ocular Diseases of Guizhou Province, Zunyi, China
| | - Qiu-Yuan Wang
- Department of Ophthalmology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Special Key Laboratory of Ocular Diseases of Guizhou Province, Zunyi, China
| | - Yang Bai
- Special Key Laboratory of Ocular Diseases of Guizhou Province, Zunyi, China
| | - Ye-Ge Bi
- Special Key Laboratory of Ocular Diseases of Guizhou Province, Zunyi, China
| | - Shan-Jun Cai
- Department of Ophthalmology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Special Key Laboratory of Ocular Diseases of Guizhou Province, Zunyi, China
| |
Collapse
|
34
|
Qi L, Heianza Y, Li X, Sacks FM, Bray GA. Toward Precision Weight-Loss Dietary Interventions: Findings from the POUNDS Lost Trial. Nutrients 2023; 15:3665. [PMID: 37630855 PMCID: PMC10458797 DOI: 10.3390/nu15163665] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 08/13/2023] [Accepted: 08/18/2023] [Indexed: 08/27/2023] Open
Abstract
The POUNDS Lost trial is a 2-year clinical trial testing the effects of dietary interventions on weight loss. This study included 811 adults with overweight or obesity who were randomized to one of four diets that contained either 15% or 25% protein and 20% or 40% fat in a 2 × 2 factorial design. By 2 years, participants on average lost from 2.9 to 3.6 kg in body weight in the four intervention arms, while no significant difference was observed across the intervention arms. In POUNDS Lost, we performed a series of ancillary studies to detect intrinsic factors particular to genomic, epigenomic, and metabolomic markers that may modulate changes in weight and other cardiometabolic traits in response to the weight-loss dietary interventions. Genomic variants identified from genome-wide association studies (GWASs) on obesity, type 2 diabetes, glucose and lipid metabolisms, gut microbiome, and dietary intakes have been found to interact with dietary macronutrients (fat, protein, and carbohydrates) in relation to weight loss and changes of body composition and cardiometabolic traits. In addition, we recently investigated epigenomic modifications, particularly blood DNA methylation and circulating microRNAs (miRNAs). We reported DNA methylation levels at NFATC2IP, CPT1A, TXNIP, and LINC00319 were related to weight loss or changes of glucose, lipids, and blood pressure; we also reported thrifty miRNA expression as a significant epigenomic marker related to changes in insulin sensitivity and adiposity. Our studies have also highlighted the importance of temporal changes in novel metabolomic signatures for gut microbiota, bile acids, and amino acids as predictors for achievement of successful weight loss outcomes. Moreover, our studies indicate that biochemical, behavioral, and psychosocial factors such as physical activity, sleep disturbance, and appetite may also modulate metabolic changes during dietary interventions. This review summarized our major findings in the POUNDS Lost trial, which provided preliminary evidence supporting the development of precision diet interventions for obesity management.
Collapse
Affiliation(s)
- Lu Qi
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA 70118, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Yoriko Heianza
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA 70118, USA
| | - Xiang Li
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA 70118, USA
| | - Frank M. Sacks
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - George A. Bray
- Department of Clinical Obesity, Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA 70808, USA
| |
Collapse
|
35
|
Nian F, Zhu C, Jin N, Xia Q, Wu L, Lu X. Gut microbiota metabolite TMAO promoted lipid deposition and fibrosis process via KRT17 in fatty liver cells in vitro. Biochem Biophys Res Commun 2023; 669:134-142. [PMID: 37271025 DOI: 10.1016/j.bbrc.2023.05.041] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 05/12/2023] [Accepted: 05/13/2023] [Indexed: 06/06/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) has become the most common chronic liver disease worldwide but still lacks specific treatment modalities. The gut microbiota and its metabolites have been shown to be intimately involved in NAFLD development, participating in and regulating disease progression. Trimethylamine N-oxide (TMAO), a metabolite highly dependent on the gut microbiota, has been shown to play deleterious regulatory roles in cardiovascular disease, but the relationship between it and NAFLD lacks validation from basic experiments. This research applied TMAO intervention by constructing fatty liver cell models in vitro to observe its effect on fatty liver cells and potential key genes and performed siRNA interference on the gene to verify the action. The results showed that TMAO intervention promoted the appearance of more red-stained lipid droplets in Oil-red O staining results, increased triglyceride (TG) levels and increased mRNA levels of liver fibrosis-related genes, and also identified one of the key genes, keratin17 (KRT17) via transcriptomics. Following the reduction in its expression level, under the same treatment, there were decreased red-stained lipid droplets, decreased TG levels, decreased indicators of impaired liver function as well as decreased mRNA levels of liver fibrosis-related genes. In conclusion, the gut microbiota metabolite TMAO could promote lipid deposition and fibrosis process via the KRT17 gene in fatty liver cells in vitro.
Collapse
Affiliation(s)
- Fulin Nian
- Department of Gastroenterology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, 2800 Gongwei Road, Pudong, Shanghai, 201399, China
| | - Chen Zhu
- Department of Gastroenterology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, 2800 Gongwei Road, Pudong, Shanghai, 201399, China
| | - Nuyun Jin
- Department of Gastroenterology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, 2800 Gongwei Road, Pudong, Shanghai, 201399, China
| | - Qiaoyun Xia
- Department of Gastroenterology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, 2800 Gongwei Road, Pudong, Shanghai, 201399, China
| | - Longyun Wu
- Department of Gastroenterology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, 2800 Gongwei Road, Pudong, Shanghai, 201399, China
| | - Xiaolan Lu
- Department of Gastroenterology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, 2800 Gongwei Road, Pudong, Shanghai, 201399, China.
| |
Collapse
|
36
|
Näätänen M, Kårlund A, Mikkonen S, Klåvus A, Savolainen O, Lehtonen M, Karhunen L, Hanhineva K, Kolehmainen M. Metabolic profiles reflect weight loss maintenance and the composition of diet after very-low-energy diet. Clin Nutr 2023; 42:1126-1141. [PMID: 37268538 DOI: 10.1016/j.clnu.2023.05.011] [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: 12/16/2022] [Revised: 05/08/2023] [Accepted: 05/12/2023] [Indexed: 06/04/2023]
Abstract
BACKGROUND & AIMS Diet and weight loss affect circulating metabolome. However, metabolite profiles induced by different weight loss maintenance diets and underlying longer term weight loss maintenance remain unknown. Herein, we investigated after-weight-loss metabolic signatures of two isocaloric 24-wk weight maintenance diets differing in satiety value due to dietary fibre, protein and fat contents and identified metabolite features that associated with successful weight loss maintenance. METHODS Non-targeted LC-MS metabolomics approach was used to analyse plasma metabolites of 79 women and men (mean age ± SD 49.7 ± 9.0 years; BMI 34.2 ± 2.5 kg/m2) participating in a weight management study. Participants underwent a 7-week very-low-energy diet (VLED) and were thereafter randomised into two groups for a 24-week weight maintenance phase. Higher satiety food (HSF) group consumed high-fibre, high-protein, and low-fat products, while lower satiety food (LSF) group consumed isocaloric low-fibre products with average protein and fat content as a part of their weight maintenance diets. Plasma metabolites were analysed before the VLED and before and after the weight maintenance phase. Metabolite features discriminating HSF and LSF groups were annotated. We also analysed metabolite features that discriminated participants who maintained ≥10% weight loss (HWM) and participants who maintained <10% weight loss (LWM) at the end of the study, irrespective of the diet. Finally, we assessed robust linear regression between metabolite features and anthropometric and food group variables. RESULTS We annotated 126 metabolites that discriminated the HSF and LSF groups and HWM and LWM groups (p < 0.05). Compared to LSF, the HSF group had lower levels of several amino acids, e.g. glutamine, arginine, and glycine, short-, medium- and long-chain acylcarnitines (CARs), odd- and even-chain lysoglycerophospholipids, and higher levels of fatty amides. Compared to LWM, the HWM group in general showed higher levels of glycerophospholipids with a saturated long-chain and a C20:4 fatty acid tail, and unsaturated free fatty acids (FFAs). Changes in several saturated odd- and even-chain LPCs and LPEs and fatty amides were associated with the intake of many food groups, particularly grain and dairy products. Increase in several (lyso)glycerophospholipids was associated with decrease in body weight and adiposity. Increased short- and medium-chain CARs were related to decreased body fat-free mass. CONCLUSIONS Our results show that isocaloric weight maintenance diets differing in dietary fibre, protein, and fat content affected amino acid and lipid metabolism. Increased abundances of several phospholipid species and FFAs were related with greater weight loss maintenance. Our findings indicate common and distinct metabolites for weight and dietary related variables in the context of weight reduction and weight management. The study was registered in isrctn.org with identifier 67529475.
Collapse
Affiliation(s)
- Mari Näätänen
- Department of Clinical Nutrition, Institute of Public Health and Clinical Nutrition, University of Eastern Finland, 70211 Kuopio, Finland.
| | - Anna Kårlund
- Department of Clinical Nutrition, Institute of Public Health and Clinical Nutrition, University of Eastern Finland, 70211 Kuopio, Finland; Department of Life Technologies, Food Sciences Unit, University of Turku, 20014 Turku, Finland.
| | - Santtu Mikkonen
- Department of Applied Physics, University of Eastern Finland, 70211 Kuopio, Finland.
| | - Anton Klåvus
- Department of Clinical Nutrition, Institute of Public Health and Clinical Nutrition, University of Eastern Finland, 70211 Kuopio, Finland.
| | - Otto Savolainen
- Department of Clinical Nutrition, Institute of Public Health and Clinical Nutrition, University of Eastern Finland, 70211 Kuopio, Finland; Department of Biology and Biological Engineering, Chalmers Mass Spectrometry Infrastructure, Chalmers University of Technology, Sweden.
| | - Marko Lehtonen
- School of Pharmacy, University of Eastern Finland, 70211 Kuopio, Finland.
| | - Leila Karhunen
- Department of Clinical Nutrition, Institute of Public Health and Clinical Nutrition, University of Eastern Finland, 70211 Kuopio, Finland.
| | - Kati Hanhineva
- Department of Clinical Nutrition, Institute of Public Health and Clinical Nutrition, University of Eastern Finland, 70211 Kuopio, Finland; Department of Life Technologies, Food Sciences Unit, University of Turku, 20014 Turku, Finland.
| | - Marjukka Kolehmainen
- Department of Clinical Nutrition, Institute of Public Health and Clinical Nutrition, University of Eastern Finland, 70211 Kuopio, Finland.
| |
Collapse
|
37
|
Li N, Li J, Wang H, Liu J, Li W, Yang K, Huo X, Leng J, Yu Z, Hu G, Fang Z, Yang X. Aromatic Amino Acids and Their Interactions with Gut Microbiota-Related Metabolites for Risk of Gestational Diabetes: A Prospective Nested Case-Control Study in a Chinese Cohort. ANNALS OF NUTRITION & METABOLISM 2023; 79:291-300. [PMID: 37339616 DOI: 10.1159/000531481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 06/05/2023] [Indexed: 06/22/2023]
Abstract
INTRODUCTION The aim of this study was to explore associations of aromatic amino acids (AAA) in early pregnancy with gestational diabetes mellitus (GDM), and whether high AAA and gut microbiota-related metabolites had interactive effects on GDM risk. METHODS We conducted a 1:1 case-control study (n = 486) nested in a prospective cohort of pregnant women from 2010 to 2012. According to the International Association of Diabetes and Pregnancy Study Group's criteria, 243 women were diagnosed with GDM. Binary conditional logistic regression was performed to examine associations of AAA with GDM risk. Interactions between AAA and gut microbiota-related metabolites for GDM were examined using additive interaction measures. RESULTS High phenylalanine and tryptophan were associated with increased GDM risk (OR: 1.72, 95% CI: 1.07-2.78 and 1.66, 1.02-2.71). The presence of high trimethylamine (TMA) markedly increased the OR of high phenylalanine alone up to 7.95 (2.79-22.71), while the presence of low glycoursodeoxycholic acid (GUDCA) markedly increased the OR of high tryptophan alone up to 22.88 (5.28-99.26), both with significant additive interactions. Furthermore, high lysophosphatidylcholines (LPC18:0) mediated both interactive effects. CONCLUSIONS High phenylalanine may have an additive interaction with high TMA, while high tryptophan may have an additive interaction with low GUDCA toward increased risk of GDM, both being mediated via LPC18:0.
Collapse
Affiliation(s)
- Ninghua Li
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Jing Li
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, China
- Tianjin Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin, China
| | - Hui Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Jinnan Liu
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Weiqin Li
- Project Office, Tianjin Women and Children's Health Center, Tianjin, China
| | - Kai Yang
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Xiaoxu Huo
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, China
- Tianjin Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin, China
| | - Junhong Leng
- Project Office, Tianjin Women and Children's Health Center, Tianjin, China
| | - Zhijie Yu
- Population Cancer Research Program and Department of Pediatrics, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Gang Hu
- Chronic Disease Epidemiology Laboratory, Pennington Biomedical Research Center, Baton Rouge, Louisiana, USA
| | - Zhongze Fang
- Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, China
- Tianjin Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin, China
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Xilin Yang
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, China
- Tianjin Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin, China
| |
Collapse
|
38
|
Liu W, Tan Z, Geng M, Jiang X, Xin Y. Impact of the gut microbiota on angiotensin Ⅱ-related disorders and its mechanisms. Biochem Pharmacol 2023:115659. [PMID: 37330020 DOI: 10.1016/j.bcp.2023.115659] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 06/08/2023] [Accepted: 06/09/2023] [Indexed: 06/19/2023]
Abstract
The renin-angiotensin system (RAS) consists of multiple angiotensin peptides and performs various biological functions mediated by distinct receptors. Angiotensin II (Ang II) is the major effector of the RAS and affects the occurrence and development of inflammation, diabetes mellitus and its complications, hypertension, and end-organ damage via the Ang II type 1 receptor. Recently, considerable interest has been given to the association and interaction between the gut microbiota and host. Increasing evidence suggests that the gut microbiota may contribute to cardiovascular diseases, obesity, type 2 diabetes mellitus, chronic inflammatory diseases, and chronic kidney disease. Recent data have confirmed that Ang II can induce an imbalance in the intestinal flora and further aggravate disease progression. Furthermore, angiotensin converting enzyme 2 is another player in RAS, alleviates the deleterious effects of Ang II, modulates gut microbial dysbiosis, local and systemic immune responses associated with coronavirus disease 19. Due to the complicated etiology of pathologies, the precise mechanisms that link disease processes with specific characteristics of the gut microbiota remain obscure. This review aims to highlight the complex interactions between the gut microbiota and its metabolites in Ang II-related disease progression, and summarize the possible mechanisms. Deciphering these mechanisms will provide a theoretical basis for novel therapeutic strategies for disease prevention and treatment. Finally, we discuss therapies targeting the gut microbiota to treat Ang II-related disorders.
Collapse
Affiliation(s)
- Wei Liu
- Key Laboratory of Pathobiology, Ministry of Education, and College of Basic Medical Sciences, Jilin University, Changchun 130021, China.
| | - Zining Tan
- Key Laboratory of Pathobiology, Ministry of Education, and College of Basic Medical Sciences, Jilin University, Changchun 130021, China.
| | - Mengrou Geng
- Key Laboratory of Pathobiology, Ministry of Education, and College of Basic Medical Sciences, Jilin University, Changchun 130021, China.
| | - Xin Jiang
- Jilin Provincial Key Laboratory of Radiation Oncology & Therapy and Department of Radiation Oncology, The First Hospital of Jilin University, Changchun 130021, China.
| | - Ying Xin
- Key Laboratory of Pathobiology, Ministry of Education, and College of Basic Medical Sciences, Jilin University, Changchun 130021, China.
| |
Collapse
|
39
|
Ai S, Li Y, Tao J, Zheng H, Tian L, Wang Y, Wang Z, Liu WJ. Bibliometric visualization analysis of gut-kidney axis from 2003 to 2022. Front Physiol 2023; 14:1176894. [PMID: 37362429 PMCID: PMC10287975 DOI: 10.3389/fphys.2023.1176894] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 05/31/2023] [Indexed: 06/28/2023] Open
Abstract
Background: The gut-kidney axis refers to the interaction between the gastrointestinal tract and the kidneys, and its disorders have become increasingly important in the development of kidney diseases. The aim of this study is to identify current research hotspots in the field of the gut-kidney axis from 2003 to 2022 and provide guidance for future research in this field. Methods: We collected relevant literature on the gut-kidney axis from the Web of Science Core Collection (WoSCC) database and conducted bibliometric and visualization analyses using biblioshiny in R-Studio and VOSviewer (version 1.6.16). Results: A total of 3,900 documents were retrieved from the WoSCC database. The publications have shown rapid expansion since 2011, with the greatest research hotspot emerging due to the concept of the "intestinal-renal syndrome," first proposed by Meijers. The most relevant journals were in the field of diet and metabolism, such as Nutrients. The United States and China were the most influential countries, and the most active institute was the University of California San Diego. Author analysis revealed that Denise Mafra, Nosratola D. Vaziri, Fouque, and Denis made great contributions in different aspects of the field. Clustering analysis of the keywords found that important research priorities were "immunity," "inflammation," "metabolism," and "urinary toxin," reflecting the basis of research in the field. Current research frontiers in the field include "hyperuricemia," "gut microbiota," "diabetes," "trimethylamine n-oxide," "iga nephropathy," "acute kidney injury," "chronic kidney disease," "inflammation," all of which necessitate further investigation. Conclusion: This study presents a comprehensive bibliometric analysis and offers an up-to-date outlook on the research related to the gut-kidney axis, with a specific emphasis on the present state of intercommunication between gut microbiota and kidney diseases in this field. This perspective may assist researchers in selecting appropriate journals and partners, and help to gain a deeper understanding of the field's hotspots and frontiers, thereby promoting future research.
Collapse
Affiliation(s)
- Sinan Ai
- Renal Research Institution of Beijing University of Chinese Medicine, Beijing, China
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yake Li
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - JiaYin Tao
- Renal Research Institution of Beijing University of Chinese Medicine, Beijing, China
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Huijuan Zheng
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Lei Tian
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Yaoxian Wang
- Renal Research Institution of Beijing University of Chinese Medicine, Beijing, China
| | - Zhen Wang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Wei Jing Liu
- Renal Research Institution of Beijing University of Chinese Medicine, Beijing, China
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
40
|
Liu Y, Zhong W, Li X, Shen F, Ma X, Yang Q, Hong S, Sun Y. Diets, Gut Microbiota and Metabolites. PHENOMICS (CHAM, SWITZERLAND) 2023; 3:268-284. [PMID: 37325710 PMCID: PMC10260722 DOI: 10.1007/s43657-023-00095-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 01/10/2023] [Accepted: 01/12/2023] [Indexed: 06/17/2023]
Abstract
The gut microbiota refers to the gross collection of microorganisms, estimated trillions of them, which reside within the gut and play crucial roles in the absorption and digestion of dietary nutrients. In the past decades, the new generation 'omics' (metagenomics, transcriptomics, proteomics, and metabolomics) technologies made it possible to precisely identify microbiota and metabolites and describe their variability between individuals, populations and even different time points within the same subjects. With massive efforts made, it is now generally accepted that the gut microbiota is a dynamically changing population, whose composition is influenced by the hosts' health conditions and lifestyles. Diet is one of the major contributors to shaping the gut microbiota. The components in the diets vary in different countries, religions, and populations. Some special diets have been adopted by people for hundreds of years aiming for better health, while the underlying mechanisms remain largely unknown. Recent studies based on volunteers or diet-treated animals demonstrated that diets can greatly and rapidly change the gut microbiota. The unique pattern of the nutrients from the diets and their metabolites produced by the gut microbiota has been linked with the occurrence of diseases, including obesity, diabetes, nonalcoholic fatty liver disease, cardiovascular disease, neural diseases, and more. This review will summarize the recent progress and current understanding of the effects of different dietary patterns on the composition of gut microbiota, bacterial metabolites, and their effects on the host's metabolism.
Collapse
Affiliation(s)
- Yilian Liu
- State Key Laboratory of Genetic Engineering and School of Life Sciences, Human Phenome Institute, Fudan University, 2005 Songhu Road, Yangpu District, Shanghai, 200433 China
| | - Wanglei Zhong
- State Key Laboratory of Genetic Engineering and School of Life Sciences, Human Phenome Institute, Fudan University, 2005 Songhu Road, Yangpu District, Shanghai, 200433 China
| | - Xiao Li
- State Key Laboratory of Genetic Engineering and School of Life Sciences, Human Phenome Institute, Fudan University, 2005 Songhu Road, Yangpu District, Shanghai, 200433 China
| | - Feng Shen
- Department of Hepatobiliary Surgery, Dongfeng Hospital, Hubei University of Medicine, Shiyan, 442001 Hubei China
| | - Xiaonan Ma
- State Key Laboratory of Genetic Engineering and School of Life Sciences, Human Phenome Institute, Fudan University, 2005 Songhu Road, Yangpu District, Shanghai, 200433 China
| | - Qi Yang
- State Key Laboratory of Genetic Engineering and School of Life Sciences, Human Phenome Institute, Fudan University, 2005 Songhu Road, Yangpu District, Shanghai, 200433 China
| | - Shangyu Hong
- State Key Laboratory of Genetic Engineering and School of Life Sciences, Human Phenome Institute, Fudan University, 2005 Songhu Road, Yangpu District, Shanghai, 200433 China
| | - Yan Sun
- Masonic Medical Research Institute, 2150 Bleecker St, Utica, NY 13501 USA
| |
Collapse
|
41
|
Bai Z, Huang X, Wu G, Ye H, Huang W, Nie Q, Chen H, Yin J, Chen Y, Nie S. Polysaccharides from red kidney bean alleviating hyperglycemia and hyperlipidemia in type 2 diabetic rats via gut microbiota and lipid metabolic modulation. Food Chem 2023; 404:134598. [DOI: 10.1016/j.foodchem.2022.134598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 09/21/2022] [Accepted: 10/10/2022] [Indexed: 11/22/2022]
|
42
|
Constantino-Jonapa LA, Espinoza-Palacios Y, Escalona-Montaño AR, Hernández-Ruiz P, Amezcua-Guerra LM, Amedei A, Aguirre-García MM. Contribution of Trimethylamine N-Oxide (TMAO) to Chronic Inflammatory and Degenerative Diseases. Biomedicines 2023; 11:biomedicines11020431. [PMID: 36830968 PMCID: PMC9952918 DOI: 10.3390/biomedicines11020431] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/23/2023] [Accepted: 01/27/2023] [Indexed: 02/05/2023] Open
Abstract
Trimethylamine N-oxide (TMAO) is a metabolite produced by the gut microbiota and has been mainly associated with an increased incidence of cardiovascular diseases (CVDs) in humans. There are factors that affect one's TMAO level, such as diet, drugs, age, and hormones, among others. Gut dysbiosis in the host has been studied recently as a new approach to understanding chronic inflammatory and degenerative diseases, including cardiovascular diseases, metabolic diseases, and Alzheimer's disease. These disease types as well as COVID-19 are known to modulate host immunity. Diabetic and obese patients have been observed to have an increase in their level of TMAO, which has a direct correlation with CVDs. This metabolite is attributed to enhancing the inflammatory pathways through cholesterol and bile acid dysregulation, promoting foam cell formation. Additionally, TMAO activates the transcription factor NF-κB, which, in turn, triggers cytokine production. The result can be an exaggerated inflammatory response capable of inducing endoplasmic reticulum stress, which is responsible for various diseases. Due to the deleterious effects that this metabolite causes in its host, it is important to search for new therapeutic agents that allow a reduction in the TMAO levels of patients and that, thus, allow patients to be able to avoid a severe cardiovascular event. The present review discussed the synthesis of TMAO and its contribution to the pathogenesis of various inflammatory diseases.
Collapse
Affiliation(s)
- Luis A. Constantino-Jonapa
- Unidad de Investigación UNAM-INC, División de Investigación, Facultad de Medicina, UNAM, Instituto Nacional de Cardiología Ignacio Chávez, Ciudad de México 14080, Mexico
| | - Yoshua Espinoza-Palacios
- Unidad de Investigación UNAM-INC, División de Investigación, Facultad de Medicina, UNAM, Instituto Nacional de Cardiología Ignacio Chávez, Ciudad de México 14080, Mexico
| | - Alma R. Escalona-Montaño
- Unidad de Investigación UNAM-INC, División de Investigación, Facultad de Medicina, UNAM, Instituto Nacional de Cardiología Ignacio Chávez, Ciudad de México 14080, Mexico
| | - Paulina Hernández-Ruiz
- Unidad de Investigación UNAM-INC, División de Investigación, Facultad de Medicina, UNAM, Instituto Nacional de Cardiología Ignacio Chávez, Ciudad de México 14080, Mexico
| | - Luis M. Amezcua-Guerra
- Departamento de Inmunología, Instituto Nacional de Cardiología Ignacio Chávez, Ciudad de México 14080, Mexico
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
- Interdisciplinary Internal Medicine Unit, Careggi University Hospital, 50134 Florence, Italy
| | - María M. Aguirre-García
- Unidad de Investigación UNAM-INC, División de Investigación, Facultad de Medicina, UNAM, Instituto Nacional de Cardiología Ignacio Chávez, Ciudad de México 14080, Mexico
- Correspondence: ; Tel.: +52-55-5573-2911 (ext. 27316)
| |
Collapse
|
43
|
Zhang N, Kandalai S, Zhou X, Hossain F, Zheng Q. Applying multi-omics toward tumor microbiome research. IMETA 2023; 2:e73. [PMID: 38868335 PMCID: PMC10989946 DOI: 10.1002/imt2.73] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 10/30/2022] [Accepted: 11/28/2022] [Indexed: 06/14/2024]
Abstract
Rather than a "short-term tenant," the tumor microbiome has been shown to play a vital role as a "permanent resident," affecting carcinogenesis, cancer development, metastasis, and cancer therapies. As the tumor microbiome has great potential to become a target for the early diagnosis and treatment of cancer, recent research on the relevance of the tumor microbiota has attracted a wide range of attention from various scientific fields, resulting in remarkable progress that benefits from the development of interdisciplinary technologies. However, there are still a great variety of challenges in this emerging area, such as the low biomass of intratumoral bacteria and unculturable character of some microbial species. Due to the complexity of tumor microbiome research (e.g., the heterogeneity of tumor microenvironment), new methods with high spatial and temporal resolution are urgently needed. Among these developing methods, multi-omics technologies (combinations of genomics, transcriptomics, proteomics, and metabolomics) are powerful approaches that can facilitate the understanding of the tumor microbiome on different levels of the central dogma. Therefore, multi-omics (especially single-cell omics) will make enormous impacts on the future studies of the interplay between microbes and tumor microenvironment. In this review, we have systematically summarized the advances in multi-omics and their existing and potential applications in tumor microbiome research, thus providing an omics toolbox for investigators to reference in the future.
Collapse
Affiliation(s)
- Nan Zhang
- Department of Radiation Oncology, College of MedicineThe Ohio State UniversityColumbusOhioUSA
- Center for Cancer Metabolism, Ohio State University Comprehensive Cancer Center ‐ James Cancer Hospital and Solove Research InstituteThe Ohio State UniversityOhioColumbusUSA
| | - Shruthi Kandalai
- Department of Radiation Oncology, College of MedicineThe Ohio State UniversityColumbusOhioUSA
- Center for Cancer Metabolism, Ohio State University Comprehensive Cancer Center ‐ James Cancer Hospital and Solove Research InstituteThe Ohio State UniversityOhioColumbusUSA
| | - Xiaozhuang Zhou
- Department of Radiation Oncology, College of MedicineThe Ohio State UniversityColumbusOhioUSA
- Center for Cancer Metabolism, Ohio State University Comprehensive Cancer Center ‐ James Cancer Hospital and Solove Research InstituteThe Ohio State UniversityOhioColumbusUSA
| | - Farzana Hossain
- Department of Radiation Oncology, College of MedicineThe Ohio State UniversityColumbusOhioUSA
- Center for Cancer Metabolism, Ohio State University Comprehensive Cancer Center ‐ James Cancer Hospital and Solove Research InstituteThe Ohio State UniversityOhioColumbusUSA
| | - Qingfei Zheng
- Department of Radiation Oncology, College of MedicineThe Ohio State UniversityColumbusOhioUSA
- Center for Cancer Metabolism, Ohio State University Comprehensive Cancer Center ‐ James Cancer Hospital and Solove Research InstituteThe Ohio State UniversityOhioColumbusUSA
- Department of Biological Chemistry and Pharmacology, College of MedicineThe Ohio State UniversityColumbusOhioUSA
| |
Collapse
|
44
|
Zhang G, Ma F, Zhang Z, Qi Z, Luo M, Yu Y. Associated long-term effects of decabromodiphenyl ethane on the gut microbial profiles and metabolic homeostasis in Sprague-Dawley rat offspring. ENVIRONMENT INTERNATIONAL 2023; 172:107802. [PMID: 36764182 DOI: 10.1016/j.envint.2023.107802] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 12/29/2022] [Accepted: 02/01/2023] [Indexed: 06/18/2023]
Abstract
Decabromodiphenyl ethane (DBDPE) as a widely used brominated flame retardant is harmful to human health due to its toxicity, including cardiovascular toxicity, reproductive toxicity, and hepatotoxicity. However, the knowledge of the long-term effects and structural and metabolic function influence on gut microbiota from DBDPE exposure remains limited. This study was mainly aimed at the gut microbiome and fecal metabolome of female rats and their offspring exposed to DBDPE in early life. 16S rRNA gene sequencing demonstrated that maternal DBDPE exposure could increase the α-diversity of gut microbiota in immature offspring while decreasing the abundance of Bifidobacterium, Clostridium, Muribaculum, Escherichia, and Lactobacillus in adult offspring. The nonmetric multidimensional scaling showed a consistency in the alternation of β-diversity between pregnant rats and their adult offspring. Furthermore, the short-chain fatty acids produced by gut microbiota dramatically increased in adult offspring after maternal DBDPE exposure, revealing that DBDPE treatment disrupted the gut microbial compositions and altered the gut community's metabolic functions. Untargeted metabolomics identified 41 differential metabolites and seven metabolic pathways between adult offspring from various groups. Targeted metabolomic showed that maternal high dose DBDPE exposure obviously decreased the level of glutathione, taurine, and l-carnitine in their adult offspring, which verified the correlation between weight loss and amino acid metabolites. An interesting link between some gut bacteria (especially the Firmicutes) and fecal metabolites demonstrated the shifts in gut microbiota may drive the metabolic process of fecal metabolites. The current findings provide new insight into long-term effects on human health.
Collapse
Affiliation(s)
- Guoxia Zhang
- NMPA Key Laboratory for Safety Evaluation of Cosmetics, Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Environmental Health, School of Public Health, Southern Medical University, Guangzhou 510515, China.
| | - Fengmin Ma
- NMPA Key Laboratory for Safety Evaluation of Cosmetics, Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Environmental Health, School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - Ziwei Zhang
- Guangdong-Hong Kong-Macao Joint Laboratory for Contaminants Exposure and Health, Guangdong Key Laboratory of Environmental Catalysis and Health Risk Control, Institute of Environmental Health and Pollution Control, Guangdong University of Technology, Guangzhou 510006, China; Guangzhou Key Laboratory of Environmental Catalysis and Pollution Control, Key Laboratory of City Cluster Environmental Safety and Green Development, School of Environmental Science and Engineering, Guangdong University of Technology, Guangzhou 510006, China
| | - Zenghua Qi
- Guangdong-Hong Kong-Macao Joint Laboratory for Contaminants Exposure and Health, Guangdong Key Laboratory of Environmental Catalysis and Health Risk Control, Institute of Environmental Health and Pollution Control, Guangdong University of Technology, Guangzhou 510006, China; Guangzhou Key Laboratory of Environmental Catalysis and Pollution Control, Key Laboratory of City Cluster Environmental Safety and Green Development, School of Environmental Science and Engineering, Guangdong University of Technology, Guangzhou 510006, China
| | - Meiqiong Luo
- Guangdong-Hong Kong-Macao Joint Laboratory for Contaminants Exposure and Health, Guangdong Key Laboratory of Environmental Catalysis and Health Risk Control, Institute of Environmental Health and Pollution Control, Guangdong University of Technology, Guangzhou 510006, China; Guangzhou Key Laboratory of Environmental Catalysis and Pollution Control, Key Laboratory of City Cluster Environmental Safety and Green Development, School of Environmental Science and Engineering, Guangdong University of Technology, Guangzhou 510006, China
| | - Yingxin Yu
- Guangdong-Hong Kong-Macao Joint Laboratory for Contaminants Exposure and Health, Guangdong Key Laboratory of Environmental Catalysis and Health Risk Control, Institute of Environmental Health and Pollution Control, Guangdong University of Technology, Guangzhou 510006, China; Guangzhou Key Laboratory of Environmental Catalysis and Pollution Control, Key Laboratory of City Cluster Environmental Safety and Green Development, School of Environmental Science and Engineering, Guangdong University of Technology, Guangzhou 510006, China.
| |
Collapse
|
45
|
The Implication of Mechanistic Approaches and the Role of the Microbiome in Polycystic Ovary Syndrome (PCOS): A Review. Metabolites 2023; 13:metabo13010129. [PMID: 36677054 PMCID: PMC9863528 DOI: 10.3390/metabo13010129] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 01/09/2023] [Accepted: 01/10/2023] [Indexed: 01/18/2023] Open
Abstract
As a complex endocrine and metabolic condition, polycystic ovarian syndrome (PCOS) affects women's reproductive health. These common symptoms include hirsutism, hyperandrogenism, ovulatory dysfunction, irregular menstruation, and infertility. No one knows what causes it or how to stop it yet. Alterations in gut microbiota composition and disruptions in secondary bile acid production appear to play a causative role in developing PCOS. PCOS pathophysiology and phenotypes are tightly related to both enteric and vaginal bacteria. Patients with PCOS exhibit changed microbiome compositions and decreased microbial diversity. Intestinal microorganisms also alter PCOS patient phenotypes by upregulating or downregulating hormone release, gut-brain mediators, and metabolite synthesis. The human body's gut microbiota, also known as the "second genome," can interact with the environment to improve metabolic and immunological function. Inflammation is connected to PCOS and may be caused by dysbiosis in the gut microbiome. This review sheds light on the recently discovered connections between gut microbiota and insulin resistance (IR) and the potential mechanisms of PCOS. This study also describes metabolomic studies to obtain a clear view of PCOS and ways to tackle it.
Collapse
|
46
|
Zhou W, Han L, Haidar Abbas Raza S, Yue Q, Sun S, Zhao Y, Lv L, Deng Y, Yuan Z, Alsharif I, Mohammedsaleh ZM, Alaryani FS, Alhumaidi Alotaibi M, Albiheyri R, Al-Sarraj F, Hasan Mukhtar M. Polysaccharides in Berberis dasystachya improve intestinal flora depending on the molecular weight and ameliorate type 2 diabetes in rats. J Funct Foods 2023. [DOI: 10.1016/j.jff.2022.105381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
|
47
|
Yan T, Shi L, Liu T, Zhang X, Yang M, Peng W, Sun X, Yan L, Dai X, Yang X. Diet-rich in wheat bran modulates tryptophan metabolism and AhR/IL-22 signalling mediated metabolic health and gut dysbacteriosis: A novel prebiotic-like activity of wheat bran. Food Res Int 2023; 163:112179. [PMID: 36596122 DOI: 10.1016/j.foodres.2022.112179] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 11/01/2022] [Accepted: 11/15/2022] [Indexed: 11/21/2022]
Abstract
Tryptophan metabolism has shown to involve in pathogenesis of various metabolic diseases. Gut microbiota-orientated diets hold great potentials to improve metabolic health via regulating tryptophan metabolism. The present study showed that the 6-week high fat diet (HFD) disturbed tryptophan metabolism accompanied with gut dysbacteriosis, also influenced the dietary tryptophan induced changes in cecum microbiome and serum metabolome in mice. The colonic expressions of aryl hydrocarbon receptor (AhR) and interleukin-22 (IL-22) were significantly reduced in mice fed on HFD. Notably, a diet- rich in wheat bran effectively inhibited transformation of tryptophan to kynurenine-pathway metabolites, while increased melatonin and microbial catabolites, i.e. indole-3-propionic acid, indole-3-acetaldehyde and 5-hydroxy-indole-3-acetic acid. Such regulatory effects were accompanied with reduced fasting glucose and total triglycerides, and promoted AhR and IL-22 levels in HFD mice. Wheat bran increased the abundance of health promoting bacteria (e.g., Akkermansia and Lactobacillus), which were significantly correlated with tryptophan derived indolic metabolites. Additionally, beneficial modulatory effects of wheat bran on indolic metabolites in associations with gut dysbacteriosis from type 2 diabetes patients were confirmed in vitro fecal fermentation experiment. Our study proves the detrimental effects of HFD induced gut dysbacteriosis on tryptophan metabolism that may influence immune modulation, and provides novel insights in the mechanisms by which wheat bran could induce health benefits.
Collapse
Affiliation(s)
- Tao Yan
- School of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Lin Shi
- School of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, Shaanxi 710119, China; Division of Food and Nutrition Science, Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg SE-412 96, Sweden.
| | - Tianqi Liu
- School of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Xiangnan Zhang
- School of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Minmin Yang
- College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Wen Peng
- Nutrition and Health Promotion Center, Department of Public Health, Medical College, Qinghai University, Xining, Qinghai 810016, China
| | - Xiaomin Sun
- Global Health Institute, Department of Nutrition and Food Safety, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China
| | - Lijing Yan
- The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Xiaoshuang Dai
- BGI Institute of Applied Agriculture, BGI-Agro, Shenzhen, Guangdong 518083, China.
| | - Xinbing Yang
- School of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, Shaanxi 710119, China.
| |
Collapse
|
48
|
Guo J, Shi CX, Zhang QQ, Deng W, Zhang LY, Chen Q, Zhang DM, Gong ZJ. Interventions for non-alcoholic liver disease: a gut microbial metabolites perspective. Therap Adv Gastroenterol 2022; 15:17562848221138676. [PMID: 36506748 PMCID: PMC9730013 DOI: 10.1177/17562848221138676] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 10/28/2022] [Indexed: 12/12/2022] Open
Abstract
Over the past two decades, non-alcoholic fatty liver disease (NAFLD) has become a leading burden of hepatocellular carcinoma and liver transplantation. Although the exact pathogenesis of NAFLD has not been fully elucidated, recent hypotheses placed more emphasis on the crucial role of the gut microbiome and its derivatives. Reportedly, microbial metabolites such as short-chain fatty acids, amino acid metabolites (indole and its derivatives), bile acids (BAs), trimethylamine N-oxide (TMAO), and endogenous ethanol exhibit sophisticated bioactive properties. These molecules regulate host lipid, glucose, and BAs metabolic homeostasis via modulating nutrient absorption, energy expenditure, inflammation, and the neuroendocrine axis. Consequently, a broad range of research has studied the therapeutic effects of microbiota-derived metabolites. In this review, we explore the interaction of microbial products and NAFLD. We also discuss the regulatory role of existing NAFLD therapies on metabolite levels and investigate the potential of targeting those metabolites to relieve NAFLD.
Collapse
Affiliation(s)
- Jin Guo
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, China
| | - Chun-Xia Shi
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qing-Qi Zhang
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, China
| | - Wei Deng
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lu-Yi Zhang
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qian Chen
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, China
| | - Dan-Mei Zhang
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, China
| | | |
Collapse
|
49
|
Yu EYW, Ren Z, Mehrkanoon S, Stehouwer CDA, van Greevenbroek MMJ, Eussen SJPM, Zeegers MP, Wesselius A. Plasma metabolomic profiling of dietary patterns associated with glucose metabolism status: The Maastricht Study. BMC Med 2022; 20:450. [PMID: 36414942 PMCID: PMC9682653 DOI: 10.1186/s12916-022-02653-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 11/07/2022] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Glucose metabolism has been reported to be affected by dietary patterns, while the underlying mechanisms involved remain unclear. This study aimed to investigate the potential mediation role of circulating metabolites in relation to dietary patterns for prediabetes and type 2 diabetes. METHODS Data was derived from The Maastricht Study that comprised of 3441 participants (mean age of 60 years) with 28% type 2 diabetes patients by design. Dietary patterns were assessed using a validated food frequency questionnaire (FFQ), and the glucose metabolism status (GMS) was defined according to WHO guidelines. Both cross-sectional and prospective analyses were performed for the circulating metabolome to investigate their associations and mediations with responses to dietary patterns and GMS. RESULTS Among 226 eligible metabolite measures obtained from targeted metabolomics, 14 were identified to be associated and mediated with three dietary patterns (i.e. Mediterranean Diet (MED), Dietary Approaches to Stop Hypertension Diet (DASH), and Dutch Healthy Diet (DHD)) and overall GMS. Of these, the mediation effects of 5 metabolite measures were consistent for all three dietary patterns and GMS. Based on a 7-year follow-up, a decreased risk for apolipoprotein A1 (APOA1) and docosahexaenoic acid (DHA) (RR 0.60, 95% CI 0.55, 0.65; RR 0.89, 95% CI 0.83, 0.97, respectively) but an increased risk for ratio of ω-6 to ω-3 fatty acids (RR 1.29, 95% CI 1.05, 1.43) of type 2 diabetes were observed from prediabetes, while APOA1 showed a decreased risk of type 2 diabetes from normal glucose metabolism (NGM; RR 0.82, 95% CI 0.75, 0.89). CONCLUSIONS In summary, this study suggests that adherence to a healthy dietary pattern (i.e. MED, DASH, or DHD) could affect the GMS through circulating metabolites, which provides novel insights into understanding the biological mechanisms of diet on glucose metabolism and leads to facilitating prevention strategy for type 2 diabetes.
Collapse
Affiliation(s)
- Evan Yi-Wen Yu
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, Department of Epidemiology & Biostatistics, School of Public Health, Southeast University, Nanjing, 210009, China. .,Department of Epidemiology, CAPHRI Care and Public Health Research Institute, Maastricht University, Universiteitssingel 40 (Room C5.570), Maastricht, 6229ER, The Netherlands.
| | - Zhewen Ren
- Department of Epidemiology, CAPHRI Care and Public Health Research Institute, Maastricht University, Universiteitssingel 40 (Room C5.570), Maastricht, 6229ER, The Netherlands
| | - Siamak Mehrkanoon
- Department of Data Science and Knowledge Engineering, Maastricht University, Maastricht, 6229ER, The Netherlands
| | - Coen D A Stehouwer
- CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, 6229ER, The Netherlands.,Department of Internal Medicine, Maastricht University Medical Center+, Maastricht, 6229HX, The Netherlands
| | - Marleen M J van Greevenbroek
- CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, 6229ER, The Netherlands.,Department of Internal Medicine, Maastricht University Medical Center+, Maastricht, 6229HX, The Netherlands
| | - Simone J P M Eussen
- Department of Epidemiology, CAPHRI Care and Public Health Research Institute, Maastricht University, Universiteitssingel 40 (Room C5.570), Maastricht, 6229ER, The Netherlands.,CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, 6229ER, The Netherlands
| | - Maurice P Zeegers
- Department of Epidemiology, CAPHRI Care and Public Health Research Institute, Maastricht University, Universiteitssingel 40 (Room C5.570), Maastricht, 6229ER, The Netherlands.,School of Nutrition and Translational Research in Metabolism, Maastricht University, Universiteitssingel 40 (Room C5.564), Maastricht, 6229ER, The Netherlands
| | - Anke Wesselius
- Department of Epidemiology, CAPHRI Care and Public Health Research Institute, Maastricht University, Universiteitssingel 40 (Room C5.570), Maastricht, 6229ER, The Netherlands. .,School of Nutrition and Translational Research in Metabolism, Maastricht University, Universiteitssingel 40 (Room C5.564), Maastricht, 6229ER, The Netherlands.
| |
Collapse
|
50
|
Canyelles M, Pérez A, Junza A, Miñambres I, Yanes O, Sardà H, Rotllan N, Julve J, Sánchez-Quesada JL, Tondo M, Escolà-Gil JC, Blanco-Vaca F. Divergent Effects of Glycemic Control and Bariatric Surgery on Circulating Concentrations of TMAO in Newly Diagnosed T2D Patients and Morbidly Obese. Diagnostics (Basel) 2022; 12:diagnostics12112783. [PMID: 36428843 PMCID: PMC9689652 DOI: 10.3390/diagnostics12112783] [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: 10/12/2022] [Revised: 11/07/2022] [Accepted: 11/10/2022] [Indexed: 11/16/2022] Open
Abstract
High circulating concentrations of the gut microbiota-derived metabolite trimethylamine N-oxide (TMAO) are significantly associated with the risk of obesity and type 2 diabetes (T2D). We aimed at evaluating the impact of glycemic control and bariatric surgery on circulating concentrations of TMAO and its microbiota-dependent intermediate, γ-butyrobetaine (γBB), in newly diagnosed T2D patients and morbidly obese subjects following a within-subject design. Based on HbA1c concentrations, T2D patients achieved glycemic control. However, the plasma TMAO and γBB concentrations were significantly increased, without changes in estimated glomerular filtration rate. Bariatric surgery was very effective in reducing weight in obese subjects. Nevertheless, the surgery reduced plasma γBB concentrations without affecting TMAO concentrations and the estimated glomerular filtration rate. Considering these results, an additional experiment was carried out in male C57BL/6J mice fed a Western-type diet for twelve weeks. Neither diet-induced obesity nor insulin resistance were associated with circulating TMAO and γBB concentrations in these genetically defined mice strains. Our findings do not support that glycemic control or bariatric surgery improve the circulating concentrations of TMAO in newly diagnosed T2D and morbidly obese patients.
Collapse
Affiliation(s)
- Marina Canyelles
- Institut de Recerca de l’Hospital Santa Creu i Sant Pau, Institut d’Investigacions Biomèdiques IIB Sant Pau, 08041 Barcelona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain
| | - Antonio Pérez
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain
- Department of Endocrinology and Nutrition, Hospital de la Santa Creu i Sant Pau, IIB Sant Pau, 08041 Barcelona, Spain
| | - Alexandra Junza
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain
- Metabolomics Platform, Department of Electronic Engineering, Universitat Rovira i Virgili, 43204 Reus, Spain
| | - Inka Miñambres
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain
- Department of Endocrinology and Nutrition, Hospital de la Santa Creu i Sant Pau, IIB Sant Pau, 08041 Barcelona, Spain
| | - Oscar Yanes
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain
- Metabolomics Platform, Department of Electronic Engineering, Universitat Rovira i Virgili, 43204 Reus, Spain
| | - Helena Sardà
- Department of Endocrinology and Nutrition, Hospital de la Santa Creu i Sant Pau, IIB Sant Pau, 08041 Barcelona, Spain
| | - Noemí Rotllan
- Institut de Recerca de l’Hospital Santa Creu i Sant Pau, Institut d’Investigacions Biomèdiques IIB Sant Pau, 08041 Barcelona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain
| | - Josep Julve
- Institut de Recerca de l’Hospital Santa Creu i Sant Pau, Institut d’Investigacions Biomèdiques IIB Sant Pau, 08041 Barcelona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain
| | - José Luis Sánchez-Quesada
- Institut de Recerca de l’Hospital Santa Creu i Sant Pau, Institut d’Investigacions Biomèdiques IIB Sant Pau, 08041 Barcelona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain
| | - Mireia Tondo
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain
- Department of Clinical Biochemistry, Hospital de la Santa Creu i Sant Pau, IIB Sant Pau, 08041 Barcelona, Spain
- Correspondence: (M.T.); (F.B.-V.)
| | - Joan Carles Escolà-Gil
- Institut de Recerca de l’Hospital Santa Creu i Sant Pau, Institut d’Investigacions Biomèdiques IIB Sant Pau, 08041 Barcelona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain
| | - Francisco Blanco-Vaca
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain
- Department of Clinical Biochemistry, Hospital de la Santa Creu i Sant Pau, IIB Sant Pau, 08041 Barcelona, Spain
- Department de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, 08041 Barcelona, Spain
- Correspondence: (M.T.); (F.B.-V.)
| |
Collapse
|