651
|
Reduced Cytokine Tumour Necrosis Factor by Pharmacological Intervention in a Preclinical Study. Biomolecules 2022; 12:biom12070877. [PMID: 35883432 PMCID: PMC9313251 DOI: 10.3390/biom12070877] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 06/15/2022] [Accepted: 06/22/2022] [Indexed: 02/04/2023] Open
Abstract
Recent preclinical studies in our laboratory have shown that the bile acid profile is altered during diabetes development and such alteration has been linked to the diabetes-associated inflammatory profile. Hence, this study aimed to investigate if the first-line antidiabetic drug metformin will alter the bile acid profile and diabetes-associated inflammation in a murine model of pre-type 2 diabetes. C57 mice were randomly allocated into three equal groups of eight. Group One was given a low-fat diet (LFD), Group Two was given a high-fat diet (HFD), and Group Three was given an HFD and, upon prediabetes confirmation, daily oral metformin for one month. Blood glucose, glycated haemoglobin, drug concentrations in tissues and faeces, and the inflammatory and bile acid profiles were measured. Metformin showed wide tissue distribution and was also present in faeces. The bile acid profile showed significant alteration due to prediabetes, and although metformin did not completely normalize it, it did exert significant effects on both the bile acid and the inflammatory profiles, suggesting a direct and, to some extent, positive impact, particularly on the diabetes-associated inflammatory profile.
Collapse
|
652
|
Gut virome profiling identifies a widespread bacteriophage family associated with metabolic syndrome. Nat Commun 2022; 13:3594. [PMID: 35739117 PMCID: PMC9226167 DOI: 10.1038/s41467-022-31390-5] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 06/14/2022] [Indexed: 11/09/2022] Open
Abstract
There is significant interest in altering the course of cardiometabolic disease development via gut microbiomes. Nevertheless, the highly abundant phage members of the complex gut ecosystem -which impact gut bacteria- remain understudied. Here, we show gut virome changes associated with metabolic syndrome (MetS), a highly prevalent clinical condition preceding cardiometabolic disease, in 196 participants by combined sequencing of bulk whole genome and virus like particle communities. MetS gut viromes exhibit decreased richness and diversity. They are enriched in phages infecting Streptococcaceae and Bacteroidaceae and depleted in those infecting Bifidobacteriaceae. Differential abundance analysis identifies eighteen viral clusters (VCs) as significantly associated with either MetS or healthy viromes. Among these are a MetS-associated Roseburia VC that is related to healthy control-associated Faecalibacterium and Oscillibacter VCs. Further analysis of these VCs revealed the Candidatus Heliusviridae, a highly widespread gut phage lineage found in 90+% of participants. The identification of the temperate Ca. Heliusviridae provides a starting point to studies of phage effects on gut bacteria and the role that this plays in MetS.
Collapse
|
653
|
Chu S, Zhang F, Wang H, Xie L, Chen Z, Zeng W, Zhou Z, Hu F. Aqueous Extract of Guava ( Psidium guajava L.) Leaf Ameliorates Hyperglycemia by Promoting Hepatic Glycogen Synthesis and Modulating Gut Microbiota. Front Pharmacol 2022; 13:907702. [PMID: 35721172 PMCID: PMC9198539 DOI: 10.3389/fphar.2022.907702] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 05/17/2022] [Indexed: 12/18/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a major global health concern. Psidium guajava L. (guava) is widely used for food as well as a folk medicine. Previous studies have shown its anti-diabetic and anti-inflammatory properties. However, the underlying mechanisms remains to be elusive. In this study, we assessed the potential therapeutic effects of aqueous extract of guava leaves (GvAEx) on T2DM and explored their potential mechanisms in vivo and in vitro. GvAEx was gavage administered for 12 weeks in diabetic db/db mice. Our results have demonstrated that GvAEx significantly lowered fasting plasma glucose levels (p < 0.01) and improved glucose tolerance and insulin sensitivity (p < 0.01, p < 0.05, respectively). Additionally, GvAEx increased hepatic glycogen accumulation, glucose uptake and decreased the mRNA expression levels of gluconeogenic genes. Furthermore, GvAEx-treatment caused higher glucose transporter 2 (GLUT2) expression in the membrane in hepatocytes. Notably, for the first time, we have elaborated the possible mechanism of the hypoglycemic effect of GvAEx from the perspective of intestinal microbiota. GvAEx has significantly changed the composition of microbiota and increased short chain fatty acid (SCFA) -producing Lachnospiraceae family and Akkermansia genus in the gut. Taken together, GvAEx could alleviate hyperglycemia and insulin resistance of T2DM by regulating glucose metabolism in the liver and restoring the gut microbiota. Thus, GvAEx has the potential for drug development against T2DM.
Collapse
Affiliation(s)
- Shuzhou Chu
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, Metabolic Syndrome Research Center, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Feng Zhang
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, Metabolic Syndrome Research Center, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Huiying Wang
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, Metabolic Syndrome Research Center, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Lijun Xie
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, Metabolic Syndrome Research Center, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Zhinan Chen
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, Metabolic Syndrome Research Center, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Weimin Zeng
- Key Laboratory of Biometallurgy, School of Minerals Processing and Bioengineering, Ministry of Education, Central South University, Changsha, China
| | - Zhiguang Zhou
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, Metabolic Syndrome Research Center, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Fang Hu
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, Metabolic Syndrome Research Center, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
654
|
Wang Y, Zhao J, Qin Y, Yu Z, Zhang Y, Ning X, Sun S. The Specific Alteration of Gut Microbiota in Diabetic Kidney Diseases—A Systematic Review and Meta-Analysis. Front Immunol 2022; 13:908219. [PMID: 35784273 PMCID: PMC9248803 DOI: 10.3389/fimmu.2022.908219] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 05/18/2022] [Indexed: 12/12/2022] Open
Abstract
Background Emerging evidence indicates that gut dysbiosis is involved in the occurrence and development of diabetic kidney diseases (DKD). However, the key microbial taxa closely related to DKD have not been determined. Methods PubMed, Web of Science, Cochrane, Chinese Biomedical Databases, China National Knowledge Internet, and Embase were searched for case-control or cross-sectional studies comparing the gut microbiota of patients with DKD and healthy controls (HC) from inception to February 8, 2022, and random/fixed-effects meta-analysis on the standardized mean difference (SMD) were performed for alpha diversity indexes between DKD and HC, and beta diversity indexes and the relative abundance of gut microbiota were extracted and summarized qualitatively. Results A total of 16 studies (578 patients with DKD and 444 HC) were included. Compared to HC, the bacterial richness of patients with DKD was significantly decreased, and the diversity indexes were decreased but not statistically, companying with a distinct beta diversity. The relative abundance of phylum Proteobacteria, Actinobacteria, and Bacteroidetes, family Coriobacteriaceae, Enterobacteriaceae, and Veillonellaceae, genus Enterococcus, Citrobacter, Escherichia, Klebsiella, Akkermansia, Sutterella, and Acinetobacter, and species E. coli were enriched while that of phylum Firmicutes, family Lachnospiraceae, genus Roseburia, Prevotella, and Bifidobacterium were depleted in patients with DKD. Conclusions The gut microbiota of patients with DKD may possess specific features characterized by expansion of genus Escherichia, Citrobacter, and Klebsiella, and depletion of Roseburia, which may contribute most to the alterations of their corresponding family and phylum taxa, as well as the bacterial diversity and composition. These microbial taxa may be closely related to DKD and serve as promising targets for the management of DKD. Systematic Review Registration https://www.crd.york.ac.uk/prospero/, identifier CRD42021289863.
Collapse
Affiliation(s)
- Yuwei Wang
- Department of Postgraduate Student, Xi’an Medical University, Xi’an, China
- Department of Nephrology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Jin Zhao
- Department of Nephrology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Yunlong Qin
- Department of Nephrology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
- Department of Nephrology, Bethune International Peace Hospital, Shijiazhuang, China
| | - Zixian Yu
- Department of Nephrology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Yumeng Zhang
- Department of Postgraduate Student, Xi’an Medical University, Xi’an, China
- Department of Nephrology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Xiaoxuan Ning
- Department of Geriatric, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Shiren Sun
- Department of Nephrology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
- *Correspondence: Shiren Sun,
| |
Collapse
|
655
|
A polysaccharide from Inonotus obliquus ameliorates intestinal barrier dysfunction in mice with type 2 diabetes mellitus. Int J Biol Macromol 2022; 214:312-323. [PMID: 35714869 DOI: 10.1016/j.ijbiomac.2022.06.071] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 06/05/2022] [Accepted: 06/10/2022] [Indexed: 12/31/2022]
Abstract
Type 2 diabetes mellitus is a global disease that endangers human health, and the need for the development of nontoxic treatment candidates is urgent. In the present work, one homogeneous polysaccharide from Inonotus obliquus (IN) was isolated, and the protective effect and mechanism of IN on type 2 diabetes mellitus were investigated from the aspects of the intestinal barrier. IN mainly consisted of 9 monosaccharides with a Mw of 373 kDa. IN attenuated body weight loss, alleviated pathological damage, and suppressed the production of proinflammatory cytokines. Additionally, IN repaired the intestinal barrier by upregulating the expression of Ki-67, ZO-1 and MUC2. Furthermore, the abundance of Firmicutes was significantly increased with IN treatment, while the levels of Bacteroidetes were significantly inhibited. In conclusion, IN protected against type 2 diabetes mellitus by ameliorating intestinal barrier dysfunction and might serve as a novel drug candidate for type 2 diabetes mellitus.
Collapse
|
656
|
Chen F, He L, Li J, Yang S, Zhang B, Zhu D, Wu Z, Zhang S, Hou D, Ouyang C, Yi J, Xiao C, Hou K. Polyethylene Glycol Loxenatide Injection (GLP-1) Protects Vascular Endothelial Cell Function in Middle-Aged and Elderly Patients With Type 2 Diabetes by Regulating Gut Microbiota. Front Mol Biosci 2022; 9:879294. [PMID: 35782875 PMCID: PMC9240776 DOI: 10.3389/fmolb.2022.879294] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Accepted: 04/19/2022] [Indexed: 02/05/2023] Open
Abstract
Objective: To evaluate the protective effect of Polyethylene Glycol Loxenatide Injection (Glucagon-like peptide-1, GLP-1) on endothelial cells from middle-aged and elderly patients with newly diagnosed or poorly controlled type 2 diabetes mellitus (T2DM). GLP-1 weekly formulation was analyzed for cardiovascular disease protection and correlated with intestinal flora. Design: Stool samples were collected from middle-aged and elderly patients with new-onset or poorly controlled type 2 diabetes in Longhu People's Hospital and Shantou Central Hospital from June 2019 to November 2019. Samples were collected at week 0, 4, and 8 of treatment with GLP-1 weekly formulations. Samples were analyzed for metagenomic sequencing. Analysis was performed to compare the characteristics of the gut microbiota at week 0, 4, and 8 of GLP-1 treatment and to correlate different microbiota with characteristic clinical parameters. Results: Statistical differences were found in blood glucose lowering, cardiovascular endothelial, and inflammation-related indices between week 0 and W4 and in blood glucose lowering and cardiovascular endothelial indices from week 0 to 8 in the newly diagnosed or poorly controlled type 2 diabetic patients treated with GLP-1. Changes in gut microbiota at week 0, 4, and 8 after using GLP-1 were not statistically different, but had an overall trend of rising and then falling, and with different bacteria, that were correlated with different clinical indicators. Conclusion: GLP-1 improves endothelial cell function indicators in middle-aged and elderly diabetic patients, which may be related to its alteration of the population numbers of gut microbiota such as Acinetobacter, Eubacterium ramulus ATCC 29099, and Bacteroides_faecis. This study provides a guidance for the treatment of type 2 diabetic patients.
Collapse
Affiliation(s)
- Fengwu Chen
- The First Affiliated Hospital of Shantou University Medical College, Shantou, China
- Department of Endocrine and Metabolic Diseases, Longhu People’s Hospital, Shantou, China
| | - Lina He
- Key Laboratory for Research on Active Ingredients in Natural Medicine of Jiangxi Province, Yichun University, Yichun, China
| | - Jilin Li
- Department of Cardiology, The Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Shuhui Yang
- Department of Endocrine and Metabolic Diseases, Shantou Central Hospital, Shantou, China
| | - Bangzhou Zhang
- School of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- School of Basic Medical Science, Central South University, Changsha, China
| | - Dan Zhu
- Department of Endocrine and Metabolic Diseases, Longhu People’s Hospital, Shantou, China
| | - Zezhen Wu
- The First Affiliated Hospital of Shantou University Medical College, Shantou, China
- Department of Endocrine and Metabolic Diseases, Longhu People’s Hospital, Shantou, China
| | - Shuo Zhang
- The First Affiliated Hospital of Shantou University Medical College, Shantou, China
- Department of Endocrine and Metabolic Diseases, Longhu People’s Hospital, Shantou, China
| | - Ducheng Hou
- Department of Endocrine and Metabolic Diseases, Longhu People’s Hospital, Shantou, China
| | - Cong Ouyang
- Center for Research and Development, Xiamen Treatgut Biotechnology Co., Ltd., Xiamen, China
| | - Jianfeng Yi
- Key Laboratory for Research on Active Ingredients in Natural Medicine of Jiangxi Province, Yichun University, Yichun, China
| | - Chuanxing Xiao
- School of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- School of Basic Medical Science, Central South University, Changsha, China
- Department of Gastroenterology, The Second Affiliated Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Kaijian Hou
- The First Affiliated Hospital of Shantou University Medical College, Shantou, China
- Department of Endocrine and Metabolic Diseases, Longhu People’s Hospital, Shantou, China
- School of Basic Medical Science, Central South University, Changsha, China
| |
Collapse
|
657
|
Plasma carnitine, choline, γ-butyrobetaine, and trimethylamine-N-oxide, but not zonulin, are reduced in overweight/obese patients with pre/diabetes or impaired glycemia. Int J Diabetes Dev Ctries 2022. [DOI: 10.1007/s13410-022-01088-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
658
|
Zhang X, Zhao A, Sandhu AK, Edirisinghe I, Burton-Freeman BM. Red Raspberry and Fructo-Oligosaccharide Supplementation, Metabolic Biomarkers, and the Gut Microbiota in Adults with Prediabetes: A Randomized Crossover Clinical Trial. J Nutr 2022; 152:1438-1449. [PMID: 35421233 DOI: 10.1093/jn/nxac037] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 09/02/2021] [Accepted: 02/14/2022] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Evidence suggests that the gut microbiota and cardiometabolic status are associated, suggesting dietary interventions that alter the microbiota may affect metabolic health. OBJECTIVES We investigated whether supplementation with (poly)phenol-dense red raspberries (RRB), alone or with a fructo-oligosaccharide (FOS) prebiotic, would improve biomarkers of cardiometabolic risk in individuals with prediabetes (PreDM) and insulin resistance (IR) and whether the effects are related to modulation of the gut microbiota. METHODS Adults with PreDM-IR (n = 26; mean ± SEM age, 35 ± 2 years; fasting glucose, 5.7 ± 0.1 mmol/L; HOMA-IR, 3.3 ± 0.3) or who were metabolically healthy (reference group; n = 10; age, 31 ± 3 years; fasting glucose, 5.1 ± 0.2 mmol/L; HOMA-IR, 1.1 ± 0.1) participated in a randomized crossover trial with two 4-week supplementation periods, in which they consumed either RRB (125 g fresh equivalents) daily or RRB + 8g FOS daily, separated by a 4-week washout. The primary outcome variable was the change in the gut microbiota composition, assessed by shotgun sequencing before (baseline) and at the end of each supplementation period. Secondary outcomes were changes in glucoregulation, lipid metabolism, anti-inflammatory status, and anthropometry. The trial is registered at ClinicalTrials.gov, NCT03049631. RESULTS In PreDM-IR, RRB supplementation reduced hepatic-IR (-30.1% ± 14.6%; P = 0.04) and reduced plasma total and LDL cholesterol [-4.9% ± 1.8% (P = 0.04) and -7.2% ± 2.3% (P = 0.003), respectively] from baseline. Adding FOS (RRB + FOS) improved β-cell function [insulin secretion rate, +70.2% ± 32.8% (P = 0.02); Disposition Index, +94.4% ± 50.2% (P = 0.04)], but had no significant effect on plasma cholesterol compared to baseline. RRB increased Eubacterium eligens (2-fold) and decreased Ruminococcus gnavus (-60% ± 34%), whereas RRB + FOS increased Bifidobacterium spp. (4-fold) and decreased Blautia wexlerae (-23% ± 12%) from baseline (all P values ≤ 0.05). R. gnavus was positively correlated with hepatic-IR, and E. eligens and Bifidobacterium catenulatum were negatively correlated with cholesterol concentrations (P ≤ 0.05). CONCLUSIONS Increased Bifidobacterium spp., concurrently with reduced R. gnavus, was associated with metabolic improvements in adults with PreDM-IR, warranting further research on the mechanisms involved in (poly)phenol/FOS-microbial interactions with host metabolism.
Collapse
Affiliation(s)
- Xuhuiqun Zhang
- Department of Food Science and Nutrition, Center for Nutrition Research and the Institute for Food Safety and Health, Illinois Institute of Technology, Chicago, IL, USA
| | - Anqi Zhao
- Department of Food Science and Nutrition, Center for Nutrition Research and the Institute for Food Safety and Health, Illinois Institute of Technology, Chicago, IL, USA
| | - Amandeep K Sandhu
- Department of Food Science and Nutrition, Center for Nutrition Research and the Institute for Food Safety and Health, Illinois Institute of Technology, Chicago, IL, USA
| | - Indika Edirisinghe
- Department of Food Science and Nutrition, Center for Nutrition Research and the Institute for Food Safety and Health, Illinois Institute of Technology, Chicago, IL, USA
| | - Britt M Burton-Freeman
- Department of Food Science and Nutrition, Center for Nutrition Research and the Institute for Food Safety and Health, Illinois Institute of Technology, Chicago, IL, USA
| |
Collapse
|
659
|
Interrelations between Gut Microbiota Composition, Nutrient Intake and Diabetes Status in an Adult Japanese Population. J Clin Med 2022; 11:jcm11113216. [PMID: 35683603 PMCID: PMC9181032 DOI: 10.3390/jcm11113216] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 05/29/2022] [Accepted: 05/30/2022] [Indexed: 01/27/2023] Open
Abstract
Upon food digestion, the gut microbiota plays a pivotal role in energy metabolism, thus affecting the development of type 2 diabetes (DM). We aimed to examine the influence of the composition of selected nutrients consumed on the association between the gut microbiota and DM. This cross-sectional study of a general population was conducted on 1019 Japanese volunteers. Compared with non-diabetic subjects, diabetic subjects had larger proportions of the genera Bifidobacterium and Streptococcus but smaller proportions of the genera Roseburia and Blautia in their gut microbiotas. The genera Streptococcus and Roseburia were positively correlated with the amounts of energy (p = 0.027) and carbohydrate and fiber (p = 0.007 and p = 0.010, respectively) consumed, respectively. In contrast, the genera Bifidobacterium and Blautia were not correlated with any of the selected nutrients consumed. Cluster analyses of these four genera revealed that the Blautia-dominant cluster was most negatively associated with DM, whereas the Bifidobacterium-dominant cluster was positively associated with DM (vs. the Blautia-dominant cluster; odds ratio 3.97, 95% confidence interval 1.68-9.35). These results indicate the possible involvement of nutrient factors in the association between the gut microbiota and DM. Furthermore, independent of nutrient factors, having a Bifidobacterium-dominant gut microbiota may be a risk factor for DM compared to having a Blautia-dominant gut microbiota in a general Japanese population.
Collapse
|
660
|
Fang X, Wu H, Wang X, Lian F, Li M, Miao R, Wei J, Tian J. Modulation of Gut Microbiota and Metabolites by Berberine in Treating Mice With Disturbances in Glucose and Lipid Metabolism. Front Pharmacol 2022; 13:870407. [PMID: 35721198 PMCID: PMC9204213 DOI: 10.3389/fphar.2022.870407] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 04/27/2022] [Indexed: 01/14/2023] Open
Abstract
Introduction: Glucose and lipid metabolism disturbances has become the third major disease after cancer and cardio-cerebrovascular diseases. Emerging evidence shows that berberine can effectively intervene glucose and lipid metabolism disturbances, but the underlying mechanisms of this remain unclear. To investigate this issue, we performed metagenomic and metabolomic analysis in a group of normal mice (the NC group), mice with disturbances in glucose and lipid metabolism (the MC group) and mice with disturbances in glucose and lipid metabolism after berberine intervention (the BER group). Result: Firstly, analysis of the clinical indicators revealed that berberine significantly improved the blood glucose and blood lipid of the host. The fasting blood glucose level decreased by approximately 30% in the BER group after 8 weeks and the oral glucose tolerance test showed that the blood glucose level of the BER group was lower than that of the MC group at any time. Besides, berberine significantly reduced body weight, total plasma cholesterol and triglyceride. Secondly, compared to the NC group, we found dramatically decreased microbial richness and diversity in the MC group and BER group. Thirdly, LDA effect size suggested that berberine significantly altered the overall gut microbiota structure and enriched many bacteria, including Akkermansia (p < 0.01), Eubacterium (p < 0.01) and Ruminococcus (p < 0.01). Fourthly, the metabolomic analysis suggested that there were significant differences in the metabolomics signature of each group. For example, isoleucine (p < 0.01), phenylalanine (p < 0.05), and arbutin (p < 0.05) significantly increased in the MC group, and berberine intervention significantly reduced them. The arbutin content in the BER group was even lower than that in the NC group. Fifthly, by combined analysis of metagenomics and metabolomics, we observed that there were significantly negative correlations between the reduced faecal metabolites (e.g., arbutin) in the BER group and the enriched gut microbiota (e.g., Eubacterium and Ruminococcus) (p < 0.05). Finally, the correlation analysis between gut microbiota and clinical indices indicated that the bacteria (e.g., Eubacterium) enriched in the BER group were negatively associated with the above-mentioned clinical indices (p < 0.05). Conclusion: Overall, our results describe that the changes of gut microbiota and metabolites are associated with berberine improving glucose and lipid metabolism disturbances.
Collapse
Affiliation(s)
- Xinyi Fang
- Department of Endocrinology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate College, Beijing University of Chinese Medicine, Beijing, China
| | - Haoran Wu
- Graduate College, Beijing University of Chinese Medicine, Beijing, China
| | - Xinmiao Wang
- Department of Endocrinology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Fengmei Lian
- Department of Endocrinology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Min Li
- Department of Endocrinology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Runyu Miao
- Department of Endocrinology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate College, Beijing University of Chinese Medicine, Beijing, China
| | - Jiahua Wei
- Graduate College, Changchun University of Chinese Medicine, Changchun, China
| | - Jiaxing Tian
- Department of Endocrinology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Jiaxing Tian,
| |
Collapse
|
661
|
Li Z, Gurung M, Rodrigues RR, Padiadpu J, Newman NK, Manes NP, Pederson JW, Greer RL, Vasquez-Perez S, You H, Hioki KA, Moulton Z, Fel A, De Nardo D, Dzutsev AK, Nita-Lazar A, Trinchieri G, Shulzhenko N, Morgun A. Microbiota and adipocyte mitochondrial damage in type 2 diabetes are linked by Mmp12+ macrophages. J Exp Med 2022; 219:213260. [PMID: 35657352 PMCID: PMC9170383 DOI: 10.1084/jem.20220017] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 03/22/2022] [Accepted: 05/05/2022] [Indexed: 01/07/2023] Open
Abstract
Microbiota contribute to the induction of type 2 diabetes by high-fat/high-sugar (HFHS) diet, but which organs/pathways are impacted by microbiota remain unknown. Using multiorgan network and transkingdom analyses, we found that microbiota-dependent impairment of OXPHOS/mitochondria in white adipose tissue (WAT) plays a primary role in regulating systemic glucose metabolism. The follow-up analysis established that Mmp12+ macrophages link microbiota-dependent inflammation and OXPHOS damage in WAT. Moreover, the molecular signature of Mmp12+ macrophages in WAT was associated with insulin resistance in obese patients. Next, we tested the functional effects of MMP12 and found that Mmp12 genetic deficiency or MMP12 inhibition improved glucose metabolism in conventional, but not in germ-free mice. MMP12 treatment induced insulin resistance in adipocytes. TLR2-ligands present in Oscillibacter valericigenes bacteria, which are expanded by HFHS, induce Mmp12 in WAT macrophages in a MYD88-ATF3-dependent manner. Thus, HFHS induces Mmp12+ macrophages and MMP12, representing a microbiota-dependent bridge between inflammation and mitochondrial damage in WAT and causing insulin resistance.
Collapse
Affiliation(s)
- Zhipeng Li
- Carlson College of Veterinary Medicine, Oregon State University, Corvallis, OR,Shanghai Mengniu Biotechnology R&D Co., Ltd., Shanghai, China
| | - Manoj Gurung
- Carlson College of Veterinary Medicine, Oregon State University, Corvallis, OR
| | - Richard R. Rodrigues
- College of Pharmacy, Oregon State University, Corvallis, OR,Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD,Basic Science Program, Frederick National Laboratory for Cancer Research, Frederick, MD
| | | | | | - Nathan P. Manes
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Jacob W. Pederson
- Carlson College of Veterinary Medicine, Oregon State University, Corvallis, OR
| | - Renee L. Greer
- Carlson College of Veterinary Medicine, Oregon State University, Corvallis, OR
| | | | - Hyekyoung You
- Carlson College of Veterinary Medicine, Oregon State University, Corvallis, OR
| | - Kaito A. Hioki
- College of Pharmacy, Oregon State University, Corvallis, OR
| | - Zoe Moulton
- Carlson College of Veterinary Medicine, Oregon State University, Corvallis, OR
| | - Anna Fel
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Dominic De Nardo
- Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Australia
| | - Amiran K. Dzutsev
- Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Aleksandra Nita-Lazar
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Giorgio Trinchieri
- Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD,Giorgio Trinchieri:
| | - Natalia Shulzhenko
- Carlson College of Veterinary Medicine, Oregon State University, Corvallis, OR,Correspondence to Natalia Shulzhenko:
| | - Andrey Morgun
- College of Pharmacy, Oregon State University, Corvallis, OR,Andrey Morgun:
| |
Collapse
|
662
|
González-Rivas JP, Pavlovska I, Polcrova A, Nieto-Martínez R, Mechanick JI. Transcultural Lifestyle Medicine in Type 2 Diabetes Care: Narrative Review of the Literature. Am J Lifestyle Med 2022. [DOI: 10.1177/15598276221095048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Disparities in type 2 diabetes (T2D) care is a global problem across diverse cultures. The Dysglycemia-Based Chronic Disease (DBCD) model promotes early and sustainable interventions along the insulin resistance (stage 1), prediabetes (stage 2), T2D (stage 3), and complications (stage 4) spectrum. In this model, lifestyle medicine is the cornerstone of preventive care to reduce DBCD progression and the socioeconomic/biological burden of disease. A comprehensive literature review, spanning 2000 to 2021, was performed and 55 studies were included examining the effects of lifestyle medicine and their cultural adaptions with different prevention modalities. In stage 1, primordial prevention targets modifiable primary drivers (behavior and environment), unhealthy lifestyles, abnormal adiposity, and insulin resistance with educational and motivational health promotion activities at individual, group, community, and population-based scales. Primary, secondary, and tertiary prevention targets individuals with mild hyperglycemia, severe hyperglycemia, and complications, respectively, using programs that incorporate structured lifestyle interventions. Culturally adapted lifestyle change in primary and secondary prevention improved quality of life and biomarkers, but with a limited impact of tertiary prevention on cardiovascular events. In conclusion, lifestyle medicine with cultural adaptations is an integral part of preventive care in patients with T2D. However, considerable research gaps exist, especially for tertiary prevention.
Collapse
Affiliation(s)
- Juan P. González-Rivas
- International Clinical Research Centre (ICRC), St Anne’s University Hospital Brno (FNUSA), Czech Republic
- Departments of Global Health and Population and Epidemiology, Harvard TH Chan School of Public Health. Harvard University, Boston, MA, USA
- Foundation for Clinic, Public Health, and Epidemiology Research of Venezuela (FISPEVEN INC), Caracas, Venezuela
| | - Iuliia Pavlovska
- International Clinical Research Centre (ICRC), St Anne’s University Hospital Brno (FNUSA), Czech Republic
- Department of Public Health, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Anna Polcrova
- International Clinical Research Centre (ICRC), St Anne’s University Hospital Brno (FNUSA), Czech Republic
- RECETOX, Faculty of Science, Masaryk University, Kotlarska 2, Brno, Czech Republic
| | - Ramfis Nieto-Martínez
- Departments of Global Health and Population and Epidemiology, Harvard TH Chan School of Public Health. Harvard University, Boston, MA, USA
- Foundation for Clinic, Public Health, and Epidemiology Research of Venezuela (FISPEVEN INC), Caracas, Venezuela
- LifeDoc Health, Memphis, TN, USA
| | - Jeffrey I. Mechanick
- he Marie-Josée and Henry R. Kravis Center for Cardiovascular Health at Mount Sinai Heart, and Division of Endocrinology, Diabetes and Bone Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
663
|
Dehghanbanadaki H, Aazami H, Ejtahed HS, Sohrabi A, Raftar SKA, Tarashi S, Tabatabaei-Malazy O, Bahramali G, Siadat SD, Esfahani EN, Razi F. The global scientific publications on gut microbiota in type 2 diabetes; a bibliometric, Scientometric, and descriptive analysis. J Diabetes Metab Disord 2022; 21:13-32. [PMID: 35673416 PMCID: PMC9167415 DOI: 10.1007/s40200-021-00920-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 10/17/2021] [Indexed: 10/19/2022]
Abstract
Background Modifying gut dysbiosis has achieved great success in managing type 2 diabetes mellitus (T2DM) and also T2DM affected the gut microbial composition. Objectives To determine the research trend of scientific publications on the relationship between gut microbiota and T2DM through a bibliometric and descriptive approach. Method We included originals and reviews related to both topics of gut microbiota and T2DM through searching in Scopus up to 31 December 2019 and then characterized their bibliometric profiles including the number of publications, citations, institutions, journals, countries, and the collaboration network of authors, countries, terms and keywords. Moreover, we performed a descriptive evaluation of the clinical trials based on their intervention type and its influence on gut dysbiosis. Results We achieved 877 articles (436 originals and 441 reviews) according to our inclusion criteria. The annual publications were constantly increased over time and reached 220 publications in 2019. Out of 436 original articles, 231 animal studies and 174 human studies were found. The majority of human studies were clinical trials (n = 77) investigating the influence of drugs (n = 21), regimens (n = 21), pre/pro/symbiotic (n = 19), surgeries (n = 15), or both drug and regimen (n = 1) on gut dysbiosis. Roux-en-Y gastric bypass and metformin were assessed the most in these trials. Obesity side by side T2DM has been assessed in this area of literature based on term and keyword analyses showing their possible similar pathways mediated by gut microbiota. Conclusion The exponentially growing documents on gut microbiota and T2DM had been published during the last decade and revealed gut microbiota alteration mediated antidiabetic effect of many interventions. Thus, we suggest other researchers to consider this pathway in efficacy assessment of therapeutic modalities and to find the optimal composition of gut microbiota that guarantees healthy insulin sensitivity. Supplementary Information The online version contains supplementary material available at 10.1007/s40200-021-00920-1.
Collapse
Affiliation(s)
- Hojat Dehghanbanadaki
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Hossein Aazami
- Metabolic Disorders Research Center, Endocrinology and Metabolism Molecular -Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Scientometrics Department, FarIdea Company, Tehran, Iran
| | - Hanieh Sadat Ejtahed
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Ahmad Sohrabi
- Cancer Control Research Center, Cancer Control Foundation, Iran University of Medical Sciences, Tehran, Iran
| | - Shahrbanoo Keshavarz Azizi Raftar
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran
- Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran
| | - Samira Tarashi
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran
- Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran
| | - Ozra Tabatabaei-Malazy
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Non-Communicable Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Golnaz Bahramali
- Hepatitis, AIDS and Bloodborne Diseases Department, Pasteur Institute of Iran, Tehran, Iran
| | - Seyed Davar Siadat
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran
- Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran
| | - Ensieh Nasli Esfahani
- Diabetes Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Farideh Razi
- Diabetes Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
664
|
Bao M, Hou K, Xin C, Zeng D, Cheng C, Zhao H, Wang Z, Wang L. Portulaca oleracea L. Extract Alleviated Type 2 Diabetes Via Modulating the Gut Microbiota and Serum Branched-Chain Amino Acid Metabolism. Mol Nutr Food Res 2022; 66:e2101030. [PMID: 35212446 DOI: 10.1002/mnfr.202101030] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Indexed: 12/21/2022]
Abstract
SCOPE Portulaca oleracea L. extracts (PE) show hypoglycemic function, but the precise mechanism remains obscure. This study is designed to investigate the association of the antidiabetes effect of PE with the gut microbiota modulation and BCAAs metabolism. METHODS AND RESULTS The Orbitrap LC-MS to Orbitrap Fusion Lumos Tribrid mass spectrometer is employed to analyze the major compounds in PE. The components of the intestinal microflora in diet-induced/STZ-treated diabetic mice are analyzed by high-throughput 16S rRNA genes sequencing. The results show that PE improves blood glucose and insulin level, increases anti-inflammatory cytokine level, lowers serum branched-chain amino acids (BCAAs), and increases serum glutamine level. PE also protects the mucosal epithelium of the colon and cecum from damage. On the impact of gut microbial composition, PE reduces the Firmicutes to Bacteroidetes ratio and the abundance of the Lachnospiraceae_NK4A136_group, Blautia, Ruminiclostridium_9, Dubosiella, and increases the abundance of the Bacteroides, Akkermansia, and Mucisprillum genera. Bacterial functionality prediction indicates PE potentially inhibits bacterial BCAAs biosynthesis, and promotes the tissue-specific expression of BCAAs catabolic enzyme for reducing BCAAs supplementation. CONCLUSION These results reveal that PE improving T2D-related biochemical abnormalities is associated not only with gut microbiota modification but also with the tissue-specific expression of BCAAs catabolic enzyme.
Collapse
Affiliation(s)
- Meili Bao
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, NO. 92 Xidazhi Street, Nangang District, Harbin, Heilongjiang, 150001, P. R. China
| | - Kexin Hou
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, NO. 92 Xidazhi Street, Nangang District, Harbin, Heilongjiang, 150001, P. R. China
| | - Chao Xin
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, NO. 92 Xidazhi Street, Nangang District, Harbin, Heilongjiang, 150001, P. R. China
| | - Deyong Zeng
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, NO. 92 Xidazhi Street, Nangang District, Harbin, Heilongjiang, 150001, P. R. China
| | - Cuilin Cheng
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, NO. 92 Xidazhi Street, Nangang District, Harbin, Heilongjiang, 150001, P. R. China
| | - Haitian Zhao
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, NO. 92 Xidazhi Street, Nangang District, Harbin, Heilongjiang, 150001, P. R. China
| | - Zhenyu Wang
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, NO. 92 Xidazhi Street, Nangang District, Harbin, Heilongjiang, 150001, P. R. China
| | - Lu Wang
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, NO. 92 Xidazhi Street, Nangang District, Harbin, Heilongjiang, 150001, P. R. China
| |
Collapse
|
665
|
Guo S, Zhang H, Chu Y, Jiang Q, Ma Y. A neural network-based framework to understand the type 2 diabetes-related alteration of the human gut microbiome. IMETA 2022; 1:e20. [PMID: 38868565 PMCID: PMC10989819 DOI: 10.1002/imt2.20] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 03/13/2022] [Accepted: 03/15/2022] [Indexed: 06/14/2024]
Abstract
The identification of microbial markers adequate to delineate the disease-related microbiome alterations from the complex human gut microbiota is of great interest. Here, we develop a framework combining neural network (NN) and random forest, resulting in 40 marker species and 90 marker genes identified from the metagenomic data set (185 healthy and 183 type 2 diabetes [T2D] samples), respectively. In terms of these markers, the NN model obtained higher accuracy in classifying the T2D-related samples than other methods; the interaction network analyses identified the key species and functional modules; the regression analysis determined that fasting blood glucose is the most significant factor (p < 0.05) in the T2D-related alteration of the human gut microbiome. We also observed that those marker species varied little across the case and control samples greatly shift in the different stages of the T2D development, suggestive of their important roles in the T2D-related microbiome alteration. Our study provides a new way of identifying the disease-related biomarkers and analyzing the role they may play in the development of the disease.
Collapse
Affiliation(s)
- Shun Guo
- Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced TechnologyChinese Academy of SciencesShenzhenGuangdongChina
- Key Laboratory of Quantitative Engineering Biology, Shenzhen Institutes of Advanced TechnologyChinese Academy of SciencesShenzhenGuangdongChina
- Shenzhen Key Laboratory of Synthetic Genomics; Guangdong Provincial Key Laboratory of Synthetic Genomics, Shenzhen Institutes of Advanced TechnologyChinese Academy of SciencesShenzhenGuangdongChina
- Shenzhen Key Lab for High Performance Data Mining, Shenzhen Institutes of Advanced TechnologyChinese Academy of SciencesShenzhenGuangdongChina
| | - Haoran Zhang
- Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced TechnologyChinese Academy of SciencesShenzhenGuangdongChina
- Key Laboratory of Quantitative Engineering Biology, Shenzhen Institutes of Advanced TechnologyChinese Academy of SciencesShenzhenGuangdongChina
- Shenzhen Key Laboratory of Synthetic Genomics; Guangdong Provincial Key Laboratory of Synthetic Genomics, Shenzhen Institutes of Advanced TechnologyChinese Academy of SciencesShenzhenGuangdongChina
| | - Yunmeng Chu
- Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced TechnologyChinese Academy of SciencesShenzhenGuangdongChina
- Key Laboratory of Quantitative Engineering Biology, Shenzhen Institutes of Advanced TechnologyChinese Academy of SciencesShenzhenGuangdongChina
- Shenzhen Key Laboratory of Synthetic Genomics; Guangdong Provincial Key Laboratory of Synthetic Genomics, Shenzhen Institutes of Advanced TechnologyChinese Academy of SciencesShenzhenGuangdongChina
| | - Qingshan Jiang
- Shenzhen Key Lab for High Performance Data Mining, Shenzhen Institutes of Advanced TechnologyChinese Academy of SciencesShenzhenGuangdongChina
| | - Yingfei Ma
- Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced TechnologyChinese Academy of SciencesShenzhenGuangdongChina
- Key Laboratory of Quantitative Engineering Biology, Shenzhen Institutes of Advanced TechnologyChinese Academy of SciencesShenzhenGuangdongChina
- Shenzhen Key Laboratory of Synthetic Genomics; Guangdong Provincial Key Laboratory of Synthetic Genomics, Shenzhen Institutes of Advanced TechnologyChinese Academy of SciencesShenzhenGuangdongChina
| |
Collapse
|
666
|
Lok KH, Wareham NJ, Nair RS, How CW, Chuah LH. Revisiting the concept of incretin and enteroendocrine L-cells as type 2 diabetes mellitus treatment. Pharmacol Res 2022; 180:106237. [PMID: 35487405 PMCID: PMC7614293 DOI: 10.1016/j.phrs.2022.106237] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 04/08/2022] [Accepted: 04/22/2022] [Indexed: 12/19/2022]
Abstract
The significant growth in type 2 diabetes mellitus (T2DM) prevalence strikes a common threat to the healthcare and economic systems globally. Despite the availability of several anti-hyperglycaemic agents in the market, none can offer T2DM remission. These agents include the prominent incretin-based therapy such as glucagon-like peptide-1 receptor (GLP-1R) agonists and dipeptidyl peptidase-4 inhibitors that are designed primarily to promote GLP-1R activation. Recent interest in various therapeutically useful gastrointestinal hormones in T2DM and obesity has surged with the realisation that enteroendocrine L-cells modulate the different incretins secretion and glucose homeostasis, reflecting the original incretin definition. Targeting L-cells offers promising opportunities to mimic the benefits of bariatric surgery on glucose homeostasis, bodyweight management, and T2DM remission. Revising the fundamental incretin theory is an essential step for therapeutic development in this area. Therefore, the present review explores enteroendocrine L-cell hormone expression, the associated nutrient-sensing mechanisms, and other physiological characteristics. Subsequently, enteroendocrine L-cell line models and the latest L-cell targeted therapies are reviewed critically in this paper. Bariatric surgery, pharmacotherapy and new paradigm of L-cell targeted pharmaceutical formulation are discussed here, offering both clinician and scientist communities a new common interest to push the scientific boundary in T2DM therapy.
Collapse
Affiliation(s)
- Kok-Hou Lok
- School of Pharmacy, Monash University Malaysia, Bandar Sunway, 47500 Subang Jaya, Selangor, Malaysia.
| | - Nicholas J Wareham
- School of Pharmacy, Monash University Malaysia, Bandar Sunway, 47500 Subang Jaya, Selangor, Malaysia; MRC Epidemiology Unit, University of Cambridge, Institute of Metabolic Science, Cambridge, UK.
| | - Rajesh Sreedharan Nair
- School of Pharmacy, Monash University Malaysia, Bandar Sunway, 47500 Subang Jaya, Selangor, Malaysia.
| | - Chee Wun How
- School of Pharmacy, Monash University Malaysia, Bandar Sunway, 47500 Subang Jaya, Selangor, Malaysia.
| | - Lay-Hong Chuah
- School of Pharmacy, Monash University Malaysia, Bandar Sunway, 47500 Subang Jaya, Selangor, Malaysia.
| |
Collapse
|
667
|
Hou K, Zhang S, Wu Z, Zhu D, Chen F, Lei ZN, Liu W, Xiao C, Chen ZS. Reconstruction of intestinal microecology of type 2 diabetes by fecal microbiota transplantation: Why and how. Bosn J Basic Med Sci 2022; 22:315-325. [PMID: 34761734 PMCID: PMC9162745 DOI: 10.17305/bjbms.2021.6323] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 10/13/2021] [Indexed: 02/05/2023] Open
Abstract
Type 2 diabetes (T2D) is a chronic metabolic disease characterized by hyperglycemia due to insulin resistance. Mounting evidence has correlated T2D to alterations in the composition of gut microbiota. Accordingly, targeting the gut microbiota has become an emerging strategy for T2D management. The aim of this article is to get a better insight into the rationale for targeting gut microbiota in T2D treatment. Thus, we herein reviewed the change of gut microbiota composition in T2D, factors shaping gut microbiota, and potential mechanisms behind the contribution of gut microbiota to T2D pathogenesis. At present, it has become possible to use intestinal microorganism capsules, bacteria liquid, and other preparations to carry out fecal microbiota transplantation for the treatment and intervention of T2D with insulin resistance and immune-mediated type 1 diabetes (T1D).
Collapse
Affiliation(s)
- Kaijian Hou
- Department of Endocrine and Metabolic Diseases, Longhu Hospital, The First Affiliated Hospital of Medical College of Shantou University, Shantou, Guangdong, China
- Department of Endocrine and Metabolic Diseases, Shantou University Medical College, Shantou University, Shantou, Guangdong, China
| | - Shuo Zhang
- Department of Endocrine and Metabolic Diseases, Longhu Hospital, The First Affiliated Hospital of Medical College of Shantou University, Shantou, Guangdong, China
- Department of Endocrine and Metabolic Diseases, Shantou University Medical College, Shantou University, Shantou, Guangdong, China
| | - Zezhen Wu
- Department of Endocrine and Metabolic Diseases, Longhu Hospital, The First Affiliated Hospital of Medical College of Shantou University, Shantou, Guangdong, China
- Department of Endocrine and Metabolic Diseases, Shantou University Medical College, Shantou University, Shantou, Guangdong, China
| | - Dan Zhu
- Department of Endocrine and Metabolic Diseases, Longhu Hospital, The First Affiliated Hospital of Medical College of Shantou University, Shantou, Guangdong, China
| | - Fengwu Chen
- Department of Endocrine and Metabolic Diseases, Longhu Hospital, The First Affiliated Hospital of Medical College of Shantou University, Shantou, Guangdong, China
| | - Zi-Ning Lei
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, New York, USA
| | - Weiting Liu
- Department of Teaching and Research Section, College of Nursing, Anhui University of Chinese Medicine, Hefei, Anhui, China
- Weiting Liu, College of Nursing, Anhui University of Chinese Medicine, No. 350, Longzihu Road, Hefei, Anhui, China
| | - Chuanxing Xiao
- Department of Pharmacy, College of Traditional Chinese Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- Corresponding authors: Chuanxing Xiao, College of Traditional Chinese Medicine, Fujian University of Traditional Chinese Medicine, No. 1, Huatuo Road, Fuzhou, Fujian, China
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, New York, USA
- Zhe-Sheng Chen, Institute for Biotechnology, St. John’s University, 8000 Utopia Parkway, Queens, New York, NY, USA
| |
Collapse
|
668
|
Qi Q, Li J, Yu B, Moon JY, Chai JC, Merino J, Hu J, Ruiz-Canela M, Rebholz C, Wang Z, Usyk M, Chen GC, Porneala BC, Wang W, Nguyen NQ, Feofanova EV, Grove ML, Wang TJ, Gerszten RE, Dupuis J, Salas-Salvadó J, Bao W, Perkins DL, Daviglus ML, Thyagarajan B, Cai J, Wang T, Manson JE, Martínez-González MA, Selvin E, Rexrode KM, Clish CB, Hu FB, Meigs JB, Knight R, Burk RD, Boerwinkle E, Kaplan RC. Host and gut microbial tryptophan metabolism and type 2 diabetes: an integrative analysis of host genetics, diet, gut microbiome and circulating metabolites in cohort studies. Gut 2022; 71:1095-1105. [PMID: 34127525 PMCID: PMC8697256 DOI: 10.1136/gutjnl-2021-324053] [Citation(s) in RCA: 138] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 06/07/2021] [Indexed: 01/05/2023]
Abstract
OBJECTIVE Tryptophan can be catabolised to various metabolites through host kynurenine and microbial indole pathways. We aimed to examine relationships of host and microbial tryptophan metabolites with incident type 2 diabetes (T2D), host genetics, diet and gut microbiota. METHOD We analysed associations between circulating levels of 11 tryptophan metabolites and incident T2D in 9180 participants of diverse racial/ethnic backgrounds from five cohorts. We examined host genome-wide variants, dietary intake and gut microbiome associated with these metabolites. RESULTS Tryptophan, four kynurenine-pathway metabolites (kynurenine, kynurenate, xanthurenate and quinolinate) and indolelactate were positively associated with T2D risk, while indolepropionate was inversely associated with T2D risk. We identified multiple host genetic variants, dietary factors, gut bacteria and their potential interplay associated with these T2D-relaetd metabolites. Intakes of fibre-rich foods, but not protein/tryptophan-rich foods, were the dietary factors most strongly associated with tryptophan metabolites. The fibre-indolepropionate association was partially explained by indolepropionate-associated gut bacteria, mostly fibre-using Firmicutes. We identified a novel association between a host functional LCT variant (determining lactase persistence) and serum indolepropionate, which might be related to a host gene-diet interaction on gut Bifidobacterium, a probiotic bacterium significantly associated with indolepropionate independent of other fibre-related bacteria. Higher milk intake was associated with higher levels of gut Bifidobacterium and serum indolepropionate only among genetically lactase non-persistent individuals. CONCLUSION Higher milk intake among lactase non-persistent individuals, and higher fibre intake were associated with a favourable profile of circulating tryptophan metabolites for T2D, potentially through the host-microbial cross-talk shifting tryptophan metabolism toward gut microbial indolepropionate production.
Collapse
Affiliation(s)
- Qibin Qi
- Department of Epidemiology and Population Health, Yeshiva University Albert Einstein College of Medicine, Bronx, New York, USA
- Department of Nutrtion, Harvard University T H Chan School of Public Health, Boston, Massachusetts, USA
| | - Jun Li
- Department of Nutrtion, Harvard University T H Chan School of Public Health, Boston, Massachusetts, USA
- Department of Epidemiology, Harvard University T H Chan School of Public Health, Boston, Massachusetts, USA
| | - Bing Yu
- Department of Epidemiology, Human Genetics, and Environmental Sciences, The University of Texas Health Science Center at Houston School of Public Health, Houston, Texas, USA
| | - Jee-Young Moon
- Department of Epidemiology and Population Health, Yeshiva University Albert Einstein College of Medicine, Bronx, New York, USA
| | - Jin C Chai
- Department of Epidemiology and Population Health, Yeshiva University Albert Einstein College of Medicine, Bronx, New York, USA
| | - Jordi Merino
- Diabetes Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Jie Hu
- Division of Women's Health, Department of Medicine at Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Miguel Ruiz-Canela
- Department of Preventive Medicine and Public Health, University of Navarra, Pamplona, Spain
- CIBER Fisiopatologıa de la Obesidad y Nutricion, Instituto de Salud Carlos III, Madrid, Spain
| | - Casey Rebholz
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Zheng Wang
- Department of Epidemiology and Population Health, Yeshiva University Albert Einstein College of Medicine, Bronx, New York, USA
| | - Mykhaylo Usyk
- Departments of Pediatrics, Microbiology and Immunology, and Gynecology and Women's Health, Yeshiva University Albert Einstein College of Medicine, Bronx, New York, USA
| | - Guo-Chong Chen
- Department of Epidemiology and Population Health, Yeshiva University Albert Einstein College of Medicine, Bronx, New York, USA
| | - Bianca C Porneala
- Division of General Internal Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Wenshuang Wang
- Department of Epidemiology, Human Genetics, and Environmental Sciences, The University of Texas Health Science Center at Houston School of Public Health, Houston, Texas, USA
- Department of Mathematics, University of Houston, Houston, Texas, USA
| | - Ngoc Quynh Nguyen
- Department of Epidemiology, Human Genetics, and Environmental Sciences, The University of Texas Health Science Center at Houston School of Public Health, Houston, Texas, USA
| | - Elena V Feofanova
- Department of Epidemiology, Human Genetics, and Environmental Sciences, The University of Texas Health Science Center at Houston School of Public Health, Houston, Texas, USA
| | - Megan L Grove
- Department of Epidemiology, Human Genetics, and Environmental Sciences, The University of Texas Health Science Center at Houston School of Public Health, Houston, Texas, USA
| | - Thomas J Wang
- Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Robert E Gerszten
- Programs in Metabolism and Medical & Population Genetics, Eli and Edythe L. Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Josée Dupuis
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, USA
| | - Jordi Salas-Salvadó
- CIBER Fisiopatologıa de la Obesidad y Nutricion, Instituto de Salud Carlos III, Madrid, Spain
- Human Nutrition Unit, Faculty of Medicine and Health Sciences, Universidad Rovira i Virgili Departamento de Medicina y Cirurgía, Reus, Spain
| | - Wei Bao
- Department of Epidemiology, The University of Iowa College of Public Health, Iowa City, Iowa, USA
| | - David L Perkins
- Institute of Minority Health Research, University of Illinois College of Medicine, Chicago, Illinois, USA
| | - Martha L Daviglus
- Institute of Minority Health Research, University of Illinois College of Medicine, Chicago, Illinois, USA
| | - Bharat Thyagarajan
- Department of Laboratory Medicine and Pathology, University of Minnesota Medical Center, Minneapolis, Minnesota, USA
| | - Jianwen Cai
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Tao Wang
- Department of Epidemiology and Population Health, Yeshiva University Albert Einstein College of Medicine, Bronx, New York, USA
| | - JoAnn E Manson
- Department of Epidemiology, Harvard University T H Chan School of Public Health, Boston, Massachusetts, USA
- Division of Preventive Medicine, Department of Medicine at Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Miguel A Martínez-González
- Department of Preventive Medicine and Public Health, University of Navarra, Pamplona, Spain
- CIBER Fisiopatologıa de la Obesidad y Nutricion, Instituto de Salud Carlos III, Madrid, Spain
| | - Elizabeth Selvin
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Kathryn M Rexrode
- Division of Women's Health, Department of Medicine at Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Clary B Clish
- Metabolomics Platform, Eli and Edythe L. Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA
| | - Frank B Hu
- Channing Division of Network Medicine, Department of Medicine at Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - James B Meigs
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
- Division of General Internal Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Rob Knight
- Department of Pediatrics, School of Medicine; Center for Microbiome Innovation, Department of Computer Science and Engineering, Jacobs School of Engineering, University of California San Diego, La Jolla, California, USA
| | - Robert D Burk
- Department of Epidemiology and Population Health, Yeshiva University Albert Einstein College of Medicine, Bronx, New York, USA
- Departments of Pediatrics, Microbiology and Immunology, and Gynecology and Women's Health, Yeshiva University Albert Einstein College of Medicine, Bronx, New York, USA
| | - Eric Boerwinkle
- Department of Epidemiology, Human Genetics, and Environmental Sciences, The University of Texas Health Science Center at Houston School of Public Health, Houston, Texas, USA
| | - Robert C Kaplan
- Department of Epidemiology and Population Health, Yeshiva University Albert Einstein College of Medicine, Bronx, New York, USA
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| |
Collapse
|
669
|
Large Yellow Tea Extract Ameliorates Metabolic Syndrome by Suppressing Lipogenesis through SIRT6/SREBP1 Pathway and Modulating Microbiota in Leptin Receptor Knockout Rats. Foods 2022; 11:foods11111638. [PMID: 35681388 PMCID: PMC9180543 DOI: 10.3390/foods11111638] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 05/30/2022] [Accepted: 05/30/2022] [Indexed: 12/12/2022] Open
Abstract
Metabolic syndrome is a chronic metabolic disorder that has turned into a severe health problem worldwide. A previous study reported that large yellow tea exhibited better anti-diabetic and lipid-lowering effects than green tea. Nevertheless, the potential mechanisms are not yet understood. In this study, we examined the prevention effects and mechanisms of large yellow tea water extract (LWE) on metabolic syndrome using leptin receptor knockout (Lepr−/−) rats. Seven-week-old male Lepr−/− and wild type (WT) littermate rats were divided into Lepr−/− control group (KO) (n = 5), Lepr−/− with LWE-treated group (KL) (n = 5), WT control group (WT) (n = 6), and WT with LWE intervention group (WL) (n = 6). Then, the rats were administered water or LWE (700 mg/kg BW) daily by oral gavage for 24 weeks, respectively. The results showed that the administration of LWE significantly reduced the serum concentrations of random blood glucose, total cholesterol, triglyceride, and free fatty acids, and increased glucose tolerance in Lepr−/− rats. Moreover, LWE remarkably reduced hepatic lipid accumulation and alleviated fatty liver formation in Lepr−/− rats. A mechanistic study showed that LWE obviously activated SIRT6 and decreased the expression of key lipogenesis-related molecules SREBP1, FAS, and DGAT1 in the livers of Lepr−/− rats. Furthermore, LWE significantly improved microbiota dysbiosis via an increase in gut microbiota diversity and an abundance of the microbiota that produce short chain fatty acids (SCFAs), such as Ruminococcaceae, Faecalibaculum, Intestinimonas, and Alistipes. Finally, LWE supplementation increased the concentrations of SCFAs in the feces of Lepr−/− rats. These results revealed that LWE attenuated metabolic syndrome of Lepr−/− rats via the reduction of hepatic lipid synthesis through the SIRT6/SREBP1 pathway and the modulation of gut microbiota.
Collapse
|
670
|
Positive effects of Epigallocatechin-3-gallate (EGCG) intervention on insulin resistance and gut microbial dysbiosis induced by bisphenol A. J Funct Foods 2022. [DOI: 10.1016/j.jff.2022.105083] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
|
671
|
Zhu M, Song Y, Martínez-Cuesta MC, Peláez C, Li E, Requena T, Wang H, Sun Y. Immunological Activity and Gut Microbiota Modulation of Pectin from Kiwano ( Cucumis metuliferus) Peels. Foods 2022; 11:foods11111632. [PMID: 35681381 PMCID: PMC9180886 DOI: 10.3390/foods11111632] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 05/21/2022] [Accepted: 05/24/2022] [Indexed: 12/22/2022] Open
Abstract
For developing the recycling of fruit by-products from kiwano, a polysaccharide was extracted from kiwano (Cucumis metuliferus) peels, namely Cucumis metuliferus peels polysaccharide (CMPP), with the aim of investigating the potential beneficial effects. The composition of polysaccharides was analyzed by chemical methods. RAW264.7 macrophages cells and the microbiota dynamics simulator (BFBL gut model) were used for in vitro study. The result showed that CMPP mainly consists of glucuronic acid, arabinose, galactose and rhamnose. By intervening with RAW264.7 cells, CMPP promoted cell proliferation and showed immune-enhancing activity, which significantly (p < 0.05) induced the release of nitric oxide (NO), tumor necrosis factor α (TNF-α) and interleukin 6 (IL-6) at a concentration of 50 μg/mL. In addition, CMPP had an impact on the composition of the gut bacteria, increasing the growth of Akkermansia, Bacteroides, Bifidobacterium, Feacalibacterium, and Roseburia. During the intake period, acetic, butyric and propionic acids were all increased, especially (p < 0.05) in the descending colon. Moreover, a decrease in ammonia concentration (10.17 ± 0.50 mM in the ascending colon, 13.21 ± 1.54 mM in the transverse colon and 13.62 ± 0.45 mM in the descending colon, respectively) was observed. In summary, CMPP can be considered as a pectin, showed immunological activity and function of gut microbiota modulation. This study could be the scientific basis of developing kiwano peels as beneficial to human health.
Collapse
Affiliation(s)
- Minqian Zhu
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou 510642, China; (M.Z.); (Y.S.); (E.L.); (Y.S.)
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Ya Song
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou 510642, China; (M.Z.); (Y.S.); (E.L.); (Y.S.)
| | - M. Carmen Martínez-Cuesta
- Department of Food Biotechnology and Microbiology, CIAL-CSIC, 28049 Madrid, Spain; (M.C.M.-C.); (C.P.)
| | - Carmen Peláez
- Department of Food Biotechnology and Microbiology, CIAL-CSIC, 28049 Madrid, Spain; (M.C.M.-C.); (C.P.)
| | - Enru Li
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou 510642, China; (M.Z.); (Y.S.); (E.L.); (Y.S.)
| | - Teresa Requena
- Department of Food Biotechnology and Microbiology, CIAL-CSIC, 28049 Madrid, Spain; (M.C.M.-C.); (C.P.)
- Correspondence: (T.R.); (H.W.)
| | - Hong Wang
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou 510642, China; (M.Z.); (Y.S.); (E.L.); (Y.S.)
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Food Science, South China Agricultural University, Guangzhou 510642, China
- Correspondence: (T.R.); (H.W.)
| | - Yuanming Sun
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou 510642, China; (M.Z.); (Y.S.); (E.L.); (Y.S.)
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| |
Collapse
|
672
|
Singh V, Park YJ, Lee G, Unno T, Shin JH. Dietary regulations for microbiota dysbiosis among post-menopausal women with type 2 diabetes. Crit Rev Food Sci Nutr 2022; 63:9961-9976. [PMID: 35635755 DOI: 10.1080/10408398.2022.2076651] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Type 2 diabetes (T2D) and T2D-associated comorbidities, such as obesity, are serious universally prevalent health issues among post-menopausal women. Menopause is an unavoidable condition characterized by the depletion of estrogen, a gonadotropic hormone responsible for secondary sexual characteristics in women. In addition to sexual dimorphism, estrogen also participates in glucose-lipid homeostasis, and estrogen depletion is associated with insulin resistance in the female body. Estrogen level in the gut also regulates the microbiota composition, and even conjugated estrogen is actively metabolized by the estrobolome to maintain insulin levels. Moreover, post-menopausal gut microbiota is different from the pre-menopausal gut microbiota, as it is less diverse and lacks the mucolytic Akkermansia and short-chain fatty acid (SCFA) producers such as Faecalibacterium and Roseburia. Through various metabolites (SCFAs, secondary bile acid, and serotonin), the gut microbiota plays a significant role in regulating glucose homeostasis, oxidative stress, and T2D-associated pro-inflammatory cytokines (IL-1, IL-6). While gut dysbiosis is common among post-menopausal women, dietary interventions such as probiotics, prebiotics, and synbiotics can ease post-menopausal gut dysbiosis. The objective of this review is to understand the relationship between post-menopausal gut dysbiosis and T2D-associated factors. Additionally, the study also provided dietary recommendations to avoid T2D progression among post-menopausal women.
Collapse
Affiliation(s)
- Vineet Singh
- Department of Applied Biosciences, Kyungpook National University, Daegu, South Korea
| | - Yeong-Jun Park
- Department of Applied Biosciences, Kyungpook National University, Daegu, South Korea
| | - GyuDae Lee
- Department of Applied Biosciences, Kyungpook National University, Daegu, South Korea
| | - Tatsuya Unno
- Department of Biotechnology, Jeju National University, Jeju, South Korea
| | - Jae-Ho Shin
- Department of Applied Biosciences, Kyungpook National University, Daegu, South Korea
| |
Collapse
|
673
|
Paul P, Kaul R, Abdellatif B, Arabi M, Upadhyay R, Saliba R, Sebah M, Chaari A. The Promising Role of Microbiome Therapy on Biomarkers of Inflammation and Oxidative Stress in Type 2 Diabetes: A Systematic and Narrative Review. Front Nutr 2022; 9:906243. [PMID: 35711547 PMCID: PMC9197462 DOI: 10.3389/fnut.2022.906243] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 04/08/2022] [Indexed: 12/12/2022] Open
Abstract
Background One in 10 adults suffer from type 2 diabetes (T2D). The role of the gut microbiome, its homeostasis, and dysbiosis has been investigated with success in the pathogenesis as well as treatment of T2D. There is an increasing volume of literature reporting interventions of pro-, pre-, and synbiotics on T2D patients. Methods Studies investigating the effect of pro-, pre-, and synbiotics on biomarkers of inflammation and oxidative stress in T2D populations were extracted from databases such as PubMed, Scopus, Web of Science, Embase, and Cochrane from inception to January 2022. Results From an initial screening of 5,984 hits, 47 clinical studies were included. Both statistically significant and non-significant results have been compiled, analyzed, and discussed. We have found various promising pro-, pre-, and synbiotic formulations. Of these, multistrain/multispecies probiotics are found to be more effective than monostrain interventions. Additionally, our findings show resistant dextrin to be the most promising prebiotic, followed closely by inulin and oligosaccharides. Finally, we report that synbiotics have shown excellent effect on markers of oxidative stress and antioxidant enzymes. We further discuss the role of metabolites in the resulting effects in biomarkers and ultimately pathogenesis of T2D, bring attention toward the ability of such nutraceuticals to have significant role in COVID-19 therapy, and finally discuss few ongoing clinical trials and prospects. Conclusion Current literature of pro-, pre- and synbiotic administration for T2D therapy is promising and shows many significant results with respect to most markers of inflammation and oxidative stress.
Collapse
Affiliation(s)
- Pradipta Paul
- Division of Medical Education, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha, Qatar
| | - Ridhima Kaul
- Division of Medical Education, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha, Qatar
| | - Basma Abdellatif
- Division of Medical Education, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha, Qatar
| | - Maryam Arabi
- Division of Premedical Education, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha, Qatar
| | - Rohit Upadhyay
- Department of Medicine—Nephrology and Hypertension, Tulane University, School of Medicine, New Orleans, LA, United States
| | - Reya Saliba
- Distributed eLibrary, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha, Qatar
| | - Majda Sebah
- Division of Premedical Education, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha, Qatar
| | - Ali Chaari
- Division of Premedical Education, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha, Qatar
| |
Collapse
|
674
|
Guo Z, Pan J, Zhu H, Chen ZY. Metabolites of Gut Microbiota and Possible Implication in Development of Diabetes Mellitus. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:5945-5960. [PMID: 35549332 DOI: 10.1021/acs.jafc.1c07851] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Diabetes mellitus is characterized by having a disorder of glucose metabolism. The types of diabetes mellitus include type 1 diabetes mellitus, type 2 diabetes mellitus, gestational diabetes mellitus, and other specific types of diabetes mellitus. Many risk factors contribute to diabetes mellitus mainly including genetics, environment, obesity, and diet. In the recent years, gut microbiota has been shown to be linked to the development of diabetes. It has been reported that the gut microbiota composition of diabetic patients is different from that of healthy people. Although the mechanism behind the abnormality remains to be explored, most hypotheses focus on the inflammation response and leaky gut in relation to the changes in production of endotoxins and metabolites derived from the intestinal flora. Consequently, the above-mentioned abnormalities trigger a series of metabolic changes, gradually leading to development of hyperglycemia, insulin resistance, and diabetes. This review is (i) to summarize the differences in gut microbiota between diabetic patients and healthy people, (ii) to discuss the underlying mechanism(s) by which how lipopolysaccharide, diet, and metabolites of the gut microbiota affect diabetes, and (iii) to provide a new insight in the prevention and treatment of diabetes.
Collapse
Affiliation(s)
- Zinan Guo
- School of Food Science and Engineering, South China Food Safety Research Center, Foshan University, Foshan 528011, Guangdong, China
- School of Life Sciences, The Chinese University of Hong Kong, Shatin 999077, Hong Kong, China
| | - Jingjin Pan
- School of Food Science and Engineering, South China Food Safety Research Center, Foshan University, Foshan 528011, Guangdong, China
| | - Hanyue Zhu
- School of Food Science and Engineering, South China Food Safety Research Center, Foshan University, Foshan 528011, Guangdong, China
| | - Zhen-Yu Chen
- School of Life Sciences, The Chinese University of Hong Kong, Shatin 999077, Hong Kong, China
| |
Collapse
|
675
|
The Impact of Air Pollution on Gut Microbiota and Children’s Health: An Expert Consensus. CHILDREN 2022; 9:children9060765. [PMID: 35740702 PMCID: PMC9222189 DOI: 10.3390/children9060765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 05/13/2022] [Accepted: 05/21/2022] [Indexed: 11/17/2022]
Abstract
Air pollution is an unseen threat to children’s health because it may increase the risk of respiratory infection, atopy, and asthma, and also alter gut microbiota compositions. The impact of air pollution on children’s health has not been firmly established. A literature review followed by a series of discussions among experts were performed to develop a theoretical framework on how air pollution could affect various bodily organs and functions in children. We invited experts from different backgrounds, such as paediatricians, nutritionists, environmental health experts, and occupational health experts, to provide their views on this matter. This report summarizes the discussion of multidisciplinary experts on the impact of air pollution on children’s health. The report begins with a review of air pollution’s impact on allergy and immunology, neurodevelopment, and cardiometabolic risks, and ends with the conceptualization of a theoretical framework. While the allergic and immunological pathway is one of the most significant pathways for air pollution affecting children’s health in which microbiotas also play a role, several pathways have been proposed regarding the ability to affect neurodevelopment and cardiometabolic risk. Further research is required to confirm the link between air pollution and the gut microbiota pathway.
Collapse
|
676
|
Jian C, Silvestre MP, Middleton D, Korpela K, Jalo E, Broderick D, de Vos WM, Fogelholm M, Taylor MW, Raben A, Poppitt S, Salonen A. Gut microbiota predicts body fat change following a low-energy diet: a PREVIEW intervention study. Genome Med 2022; 14:54. [PMID: 35599315 PMCID: PMC9125896 DOI: 10.1186/s13073-022-01053-7] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 05/04/2022] [Indexed: 12/17/2022] Open
Abstract
Background Low-energy diets (LEDs) comprise commercially formulated food products that provide between 800 and 1200 kcal/day (3.3–5 MJ/day) to aid body weight loss. Recent small-scale studies suggest that LEDs are associated with marked changes in the gut microbiota that may modify the effect of the LED on host metabolism and weight loss. We investigated how the gut microbiota changed during 8 weeks of total meal replacement LED and determined their associations with host response in a sub-analysis of 211 overweight adults with pre-diabetes participating in the large multicentre PREVIEW (PREVention of diabetes through lifestyle intervention and population studies In Europe and around the World) clinical trial. Methods Microbial community composition was analysed by Illumina sequencing of the hypervariable V3-V4 regions of the 16S ribosomal RNA (rRNA) gene. Butyrate production capacity was estimated by qPCR targeting the butyryl-CoA:acetate CoA-transferase gene. Bioinformatics and statistical analyses, such as comparison of alpha and beta diversity measures, correlative and differential abundances analysis, were undertaken on the 16S rRNA gene sequences of 211 paired (pre- and post-LED) samples as well as their integration with the clinical, biomedical and dietary datasets for predictive modelling. Results The overall composition of the gut microbiota changed markedly and consistently from pre- to post-LED (P = 0.001), along with increased richness and diversity (both P < 0.001). Following the intervention, the relative abundance of several genera previously associated with metabolic improvements (e.g., Akkermansia and Christensenellaceae R-7 group) was significantly increased (P < 0.001), while flagellated Pseudobutyrivibrio, acetogenic Blautia and Bifidobacterium spp. were decreased (all P < 0.001). Butyrate production capacity was reduced (P < 0.001). The changes in microbiota composition and predicted functions were significantly associated with body weight loss (P < 0.05). Baseline gut microbiota features were able to explain ~25% of variation in total body fat change (post–pre-LED). Conclusions The gut microbiota and individual taxa were significantly influenced by the LED intervention and correlated with changes in total body fat and body weight in individuals with overweight and pre-diabetes. Despite inter-individual variation, the baseline gut microbiota was a strong predictor of total body fat change during the energy restriction period. Trial registration The PREVIEW trial was prospectively registered at ClinicalTrials.gov (NCT01777893) on January 29, 2013. Supplementary Information The online version contains supplementary material available at 10.1186/s13073-022-01053-7.
Collapse
|
677
|
Aggarwal V, Sunder S, Verma SR. Disease-associated dysbiosis and potential therapeutic role of Akkermansia muciniphila, a mucus degrading bacteria of gut microbiome. Folia Microbiol (Praha) 2022; 67:811-824. [PMID: 35596115 PMCID: PMC9122250 DOI: 10.1007/s12223-022-00973-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Accepted: 04/19/2022] [Indexed: 02/08/2023]
Abstract
The unique functionality of Akkermansia muciniphila in gut microbiota indicates it to be an indispensable microbe for human welfare. The importance of A. muciniphila lies in its potential to convert mucin into beneficial by-products, regulate intestinal homeostasis and maintain gut barrier integrity. It is also known to competitively inhibit other mucin-degrading bacteria and improve metabolic functions and immunity responses in the host. It finds a pivotal perspective in various diseases and their treatment. It has future as a promising probiotic, disease biomarker and therapeutic agent for chronic diseases. Disease-associated dysbiosis of A. muciniphila in the gut microbiome makes it a potential candidate as a biomarker for some diseases and can provide future theranostics by suggesting ways of diagnosis for the patients and best treatment method based on the screening results. Manipulation of A. muciniphila in gut microbiome may help in developing a novel personalized therapeutic action and can be a suitable next generation medicine. However, the actual pathway governing A. muciniphila interaction with hosts remains to be investigated. Also, due to the limited availability of products containing A. muciniphila, it is not exploited to its full potential. The present review aims at highlighting the potential of A. muciniphila in mucin degradation, contribution towards the gut health and host immunity and management of metabolic diseases such as obesity and type 2 diabetes, and respiratory diseases such as cystic fibrosis and COVID-19.
Collapse
Affiliation(s)
- Vidushi Aggarwal
- Department of Biotechnology, Delhi Technological University, Delhi, 110042, India
| | - Sushant Sunder
- Department of Biotechnology, Delhi Technological University, Delhi, 110042, India
| | - Smita Rastogi Verma
- Department of Biotechnology, Delhi Technological University, Delhi, 110042, India.
| |
Collapse
|
678
|
Park J, Hosomi K, Kawashima H, Chen YA, Mohsen A, Ohno H, Konishi K, Tanisawa K, Kifushi M, Kogawa M, Takeyama H, Murakami H, Kubota T, Miyachi M, Kunisawa J, Mizuguchi K. Dietary Vitamin B1 Intake Influences Gut Microbial Community and the Consequent Production of Short-Chain Fatty Acids. Nutrients 2022; 14:nu14102078. [PMID: 35631219 PMCID: PMC9147846 DOI: 10.3390/nu14102078] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 05/09/2022] [Accepted: 05/13/2022] [Indexed: 12/11/2022] Open
Abstract
The gut microbiota is closely related to good health; thus, there have been extensive efforts dedicated to improving health by controlling the gut microbial environment. Probiotics and prebiotics are being developed to support a healthier intestinal environment. However, much work remains to be performed to provide effective solutions to overcome individual differences in the gut microbial community. This study examined the importance of nutrients, other than dietary fiber, on the survival of gut bacteria in high-health-conscious populations. We found that vitamin B1, which is an essential nutrient for humans, had a significant effect on the survival and competition of bacteria in the symbiotic gut microbiota. In particular, sufficient dietary vitamin B1 intake affects the relative abundance of Ruminococcaceae, and these bacteria have proven to require dietary vitamin B1 because they lack the de novo vitamin B1 synthetic pathway. Moreover, we demonstrated that vitamin B1 is involved in the production of butyrate, along with the amount of acetate in the intestinal environment. We established the causality of possible associations and obtained mechanical insight, through in vivo murine experiments and in silico pathway analyses. These findings serve as a reference to support the development of methods to establish optimal intestinal environment conditions for healthy lifestyles.
Collapse
Affiliation(s)
- Jonguk Park
- Artificial Intelligence Center for Health and Biomedical Research, National Institutes of Biomedical Innovation, Health and Nutrition, 7-6-8 Saito-Asagi, Osaka 567-0085, Ibaraki, Japan; (H.K.); (Y.-A.C.); (A.M.)
- Correspondence: (J.P.); (J.K.); (K.M.)
| | - Koji Hosomi
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research and Laboratory of Gut Environmental System, National Institutes of Biomedical Innovation, Health and Nutrition, 7-6-8 Saito-Asagi, Osaka 567-0085, Ibaraki, Japan;
| | - Hitoshi Kawashima
- Artificial Intelligence Center for Health and Biomedical Research, National Institutes of Biomedical Innovation, Health and Nutrition, 7-6-8 Saito-Asagi, Osaka 567-0085, Ibaraki, Japan; (H.K.); (Y.-A.C.); (A.M.)
| | - Yi-An Chen
- Artificial Intelligence Center for Health and Biomedical Research, National Institutes of Biomedical Innovation, Health and Nutrition, 7-6-8 Saito-Asagi, Osaka 567-0085, Ibaraki, Japan; (H.K.); (Y.-A.C.); (A.M.)
| | - Attayeb Mohsen
- Artificial Intelligence Center for Health and Biomedical Research, National Institutes of Biomedical Innovation, Health and Nutrition, 7-6-8 Saito-Asagi, Osaka 567-0085, Ibaraki, Japan; (H.K.); (Y.-A.C.); (A.M.)
| | - Harumi Ohno
- Department of Physical Activity Research, National Institutes of Biomedical Innovation, Health and Nutrition, 1-23-1 Toyama, Shinjuku 162-8636, Tokyo, Japan or (H.O.); (K.K.); (K.T.); (H.M.); (M.M.)
- Department of Nutrition, Kiryu University, 606-7 Azami, Kasakake-machi, Midori 379-2392, Gunma, Japan
| | - Kana Konishi
- Department of Physical Activity Research, National Institutes of Biomedical Innovation, Health and Nutrition, 1-23-1 Toyama, Shinjuku 162-8636, Tokyo, Japan or (H.O.); (K.K.); (K.T.); (H.M.); (M.M.)
- Faculty of Food and Nutritional Sciences, Toyo University, 1-1-1 Izumino, Itakura, Oura 374-0193, Gunma, Japan
| | - Kumpei Tanisawa
- Department of Physical Activity Research, National Institutes of Biomedical Innovation, Health and Nutrition, 1-23-1 Toyama, Shinjuku 162-8636, Tokyo, Japan or (H.O.); (K.K.); (K.T.); (H.M.); (M.M.)
- School of Sport Sciences, Waseda University, 2-579-15 Mikajima, Tokorozawa 359-1192, Saitama, Japan
| | - Masako Kifushi
- Department of Life Science and Medical Bioscience, Graduate School of Advanced Science and Engineering, Waseda University, 2-2 Wakamatsucho, Shinjuku 162-8480, Tokyo, Japan; (M.K.); (H.T.)
- Computational Bio Big-Data Open Innovation Laboratory, National Institute of Advanced Industrial Science and Technology, 3-4-1 Okubo, Shinjuku 169-8555, Tokyo, Japan
| | - Masato Kogawa
- Research Organization for Nano and Life Innovation, Waseda University, 513 Wasedatsurumaki, Shinjuku 162-0041, Tokyo, Japan;
| | - Haruko Takeyama
- Department of Life Science and Medical Bioscience, Graduate School of Advanced Science and Engineering, Waseda University, 2-2 Wakamatsucho, Shinjuku 162-8480, Tokyo, Japan; (M.K.); (H.T.)
- Computational Bio Big-Data Open Innovation Laboratory, National Institute of Advanced Industrial Science and Technology, 3-4-1 Okubo, Shinjuku 169-8555, Tokyo, Japan
- Research Organization for Nano and Life Innovation, Waseda University, 513 Wasedatsurumaki, Shinjuku 162-0041, Tokyo, Japan;
- Institute for Advanced Research of Biosystem Dynamics, Waseda Research Institute for Science and Engineering, Graduate School of Advanced Science and Engineering, Waseda University, 3-4-1 Okubo, Shinjuku 169-8555, Tokyo, Japan
| | - Haruka Murakami
- Department of Physical Activity Research, National Institutes of Biomedical Innovation, Health and Nutrition, 1-23-1 Toyama, Shinjuku 162-8636, Tokyo, Japan or (H.O.); (K.K.); (K.T.); (H.M.); (M.M.)
- Faculty of Sport and Health Science, Ritsumeikan University, 1-1-1 Nojihigashi, Kusatsu 525-0085, Shiga, Japan
| | - Tetsuya Kubota
- Department of Clinical Nutrition, National Institutes of Biomedical Innovation, Health and Nutrition, 1-23-1 Toyama, Shinjuku 162-8636, Tokyo, Japan;
- Laboratory for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences, 1-7-22 Suehiro, Tsurumi, Yokohama 230-0045, Kanagawa, Japan
- Division of Diabetes and Metabolism, The Institute for Medical Science, Asahi Life Foundation, 2-2-6 Nihonbashibakuro, Chuo 103-0002, Tokyo, Japan
| | - Motohiko Miyachi
- Department of Physical Activity Research, National Institutes of Biomedical Innovation, Health and Nutrition, 1-23-1 Toyama, Shinjuku 162-8636, Tokyo, Japan or (H.O.); (K.K.); (K.T.); (H.M.); (M.M.)
- School of Sport Sciences, Waseda University, 2-579-15 Mikajima, Tokorozawa 359-1192, Saitama, Japan
| | - Jun Kunisawa
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research and Laboratory of Gut Environmental System, National Institutes of Biomedical Innovation, Health and Nutrition, 7-6-8 Saito-Asagi, Osaka 567-0085, Ibaraki, Japan;
- Research Organization for Nano and Life Innovation, Waseda University, 513 Wasedatsurumaki, Shinjuku 162-0041, Tokyo, Japan;
- International Research and Development Center for Mucosal Vaccines, Institute of Medical Science, University of Tokyo, 4-6-1 Shirokanedai, Minato 108-8639, Tokyo, Japan
- Graduate School of Medicine, Graduate School of Pharmaceutical Sciences, Graduate School of Dentistry, Graduate School of Sciences, Osaka University, 1-1 Yamadaoka, Suita 565-0871, Osaka, Japan
- Department of Microbiology and Immunology, Graduate School of Medicine, Kobe University, 7-5-1 Kusunoki, Chuo, Kobe 650-0017, Hyogo, Japan
- Correspondence: (J.P.); (J.K.); (K.M.)
| | - Kenji Mizuguchi
- Artificial Intelligence Center for Health and Biomedical Research, National Institutes of Biomedical Innovation, Health and Nutrition, 7-6-8 Saito-Asagi, Osaka 567-0085, Ibaraki, Japan; (H.K.); (Y.-A.C.); (A.M.)
- Institute for Protein Research, Osaka University, 3-2 Yamadaoka, Suita 565-0871, Osaka, Japan
- Correspondence: (J.P.); (J.K.); (K.M.)
| |
Collapse
|
679
|
Ren X, Xing Y, He L, Xiu Z, Yang L, Han A, Jia Q, Dong Y. Effect of 1-Deoxynojirimycin on insulin resistance in prediabetic mice based on next-generation sequencing and intestinal microbiota study. JOURNAL OF ETHNOPHARMACOLOGY 2022; 289:115029. [PMID: 35077826 DOI: 10.1016/j.jep.2022.115029] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 01/14/2022] [Accepted: 01/20/2022] [Indexed: 06/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE 1-Deoxynojirimycin (DNJ), the major alkaloid in Morus alba L., is the main effective constituent in "Mulberry twig Alkaloids Tablets" launched in China in 2020. Prediabetes, characterized by insulin resistance, is regarded as the key period for reversing Type 2 diabetes mellitus (T2DM) through lifestyle intervention and glucose-lowering drugs. Besides the excellent activity as an α-glucosidase inhibitor, DNJ also improves insulin sensitivity in T2DM murine models, yet the mechanism is still unclear. Besides, the pharmaceutical effect of DNJ on prediabetes is also undocumented. AIM OF THE STUDY The aim of this study was to investigate the pharmaceutical effect of DNJ on high-fat and streptozotocin (STZ)-induced prediabetes mice, and to elucidate the mechanism of insulin resistance ameliorated by DNJ. MATERIALS AND METHODS Oral glucose tolerance test (OGTT) and insulin tolerance test (ITT) were performed to detect blood glucose level and insulin sensitivity in mice. The levels of circulating lipopolysaccharide (LPS), interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α) in the plasma of mice were measured by limulus reagent and enzyme-linked immunosorbent assay (ELISA), respectively. Next-generation sequencing (NGS) and intestinal microbiota sequencing were used to screen the alterations in the transcriptome of liver tissues and to assess the differences in intestinal flora composition, respectively. Expression of cytokine signaling pathway inhibitor 3 (SOCS3), insulin receptor substrate (IRS1), p-IRS1 (Tyr896), occludin, and toll like receptor 4 (TLR4)/NF-κB signaling pathway were confirmed by western blotting. RESULTS Our study revealed that DNJ decreased the blood glucose level and improve insulin sensitivity in prediabetic mice. DNJ significantly reduced the relative risk of T2DM in prediabetic mice by approximately 83.7%. Mechanistically, DNJ treatment suppressed the circulating levels of LPS, IL-6, and TNF-α in plasma and decreased the inflammatory infiltration in liver and colon tissues. DNJ-treatment increased the abundance of Akkermansia, Bifidobacterium, and Lactobacillus, and decreased the abundance of Enterococcaceae and Lachnospiraceae. Moreover, DNJ suppressed the expression of SOCS3 and the activity of TLR4/NF-κB signaling pathway, meanwhile improving the expression of occludin and the ratio of p-IRS1 (Tyr896)/IRS1. CONCLUSIONS DNJ effectively ameliorates glucose and lipid metabolism in prediabetic mice, and decreased the relative risk of progression into T2DM from prediabetes. The suppressed immune responses play essential roles in the improvement of insulin resistance by DNJ treatment. In conclusion, DNJ from Morus alba L. is a promising alternative agent in T2DM prevention.
Collapse
Affiliation(s)
- Xinxiu Ren
- School of Bioengineering, Dalian University of Technology, Dalian, Liaoning, 116024, China.
| | - Yan Xing
- School of Bioengineering, Dalian University of Technology, Dalian, Liaoning, 116024, China.
| | - Liangyu He
- School of Bioengineering, Dalian University of Technology, Dalian, Liaoning, 116024, China.
| | - Zhilong Xiu
- School of Bioengineering, Dalian University of Technology, Dalian, Liaoning, 116024, China.
| | - Ling Yang
- Xinjiang Production and Construction Corps Key Laboratory of Protection and Utilization of Biological Resources in Tarim Basin, Tarim University, Alar, Xinjiang, 843300, China; Instrumental Analysis Center, Tarim University, Alar, Xinjiang, 843300, China.
| | - Aizhi Han
- Instrumental Analysis Center, Tarim University, Alar, Xinjiang, 843300, China.
| | - Qinhua Jia
- Instrumental Analysis Center, Tarim University, Alar, Xinjiang, 843300, China.
| | - Yuesheng Dong
- School of Bioengineering, Dalian University of Technology, Dalian, Liaoning, 116024, China.
| |
Collapse
|
680
|
Jung W, Jang S. Oral Microbiome Research on Oral Lichen Planus: Current Findings and Perspectives. BIOLOGY 2022; 11:biology11050723. [PMID: 35625451 PMCID: PMC9138428 DOI: 10.3390/biology11050723] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 05/05/2022] [Accepted: 05/07/2022] [Indexed: 12/12/2022]
Abstract
Simple Summary Oral lichen planus is a disease of the oral mucosa, which frequently affects women aged 40 years or older. Though the T cell-mediated immune response is involved in the development of oral lichen planus, attempts to identify a microorganism that causes the disease have been unsuccessful. Recent studies on the development of oral lichen planus are focusing on the role of the oral microbiome, which includes oral microbiota and their products, and the host environment. The role of the human microbiome in various diseases has been identified and regulating the microbiome is becoming important in personalized medicine. In this review, we summarized current findings on the role of the oral microbiome in the development of oral lichen planus. The homeostasis of the oral microbiome is disrupted in patients, and functional analysis of oral microbiota and oral mucosa implies that pathways involved in defense against bacterial infection and in the inflammatory response are activated in the oral lichen planus-associated oral microbiome. Though the lack of studies to date makes it difficult to conclude, further studies on the oral microbiome associated with the disease will enable a holistic understanding of the role of the oral microbiome in the development of oral lichen planus and developing a personalized therapy for the disease. Abstract Oral lichen planus (OLP) is a chronic inflammatory disease of the oral mucosa with an unknown etiology. The role of oral microbes in the development of OLP has gained researchers’ interest. In this review, we summarized the findings of studies focused on the relationship between OLP and oral microbiome, which includes the composition of oral microbiota, molecules produced by oral microbiota or the host, and the oral environment of the host. According to the studies, the oral microbial community in OLP patients undergoes dysbiosis, and the microbial dysbiosis in OLP patients is more prominent in the buccal mucosa than in the saliva. However, no same microorganisms have been suggested to be associated with OLP in multiple investigations, implying that the functional aspects of the oral microbiota are more important in OLP development than the composition of the oral microbiota. According to studies on host factors that make up the oral environment, signal pathways involved in cellular processes, such as keratinization, inflammation, and T cell responses are triggered in OLP. Studies on the functional aspects of the oral microbiota, as well as interactions between the host and the oral microbiota, are still lacking, and more research is required.
Collapse
Affiliation(s)
- Won Jung
- Department of Oral Medicine, Institute of Oral Bioscience, School of Dentistry, Jeonbuk National University, Jeonju-si 54907, Korea;
- Research Institute of Clinical Medicine, Jeonbuk National University, Jeonju-si 54907, Korea
- Biomedical Research Institute, Jeonbuk National University Hospital, Jeonju-si 54907, Korea
| | - Sungil Jang
- Department of Oral Biochemistry, Institute of Oral Bioscience, School of Dentistry, Jeonbuk National University, Jeonju-si 54907, Korea
- Correspondence: ; Tel.: +82-63-270-4027
| |
Collapse
|
681
|
Meleshko T, Rukavchuk R, Levchuk O, Boyko N. Personalized Nutrition for Microbiota Correction and Metabolism Restore in Type 2 Diabetes Mellitus Patients. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1369:1-16. [PMID: 33634376 DOI: 10.1007/5584_2021_621] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Type 2 diabetes is one of the most common noncommunicable diseases in the world. Recent studies suggest a link between type 2 diabetes and microbiota, as well as the ability to treat and prevent it using personalized approaches to nutrition. In this work, we conducted clinical studies on the effects of a personalized diet on 56 female patients. Biochemical, physical, and immunological parameters were measured by standard methods on days 1 and 18 of the experiment. Gut and oral microbiota studies were performed in dynamics on days 1, 7, 11, and 18 using real-time polymerase chain reaction. With the help of the developed information system, a personalized diet was developed for each participant of the experiment. In the group of patients following personalized diets a statistically significant decreasing levels of glucose, thymol test, creatinine, very low-density lipoprotein, urea, secretory IgA, and tumour necrosis factor-α, and improvement in all physical parameters were observed. There was a statistically significant increase in uric acid, sodium, and magnesium. Statistically significant changes in gut microbiota were observed in Enterococcus faecalis, Escherichia coli (lac+, lac-), Lactobacillus spp., and Candida spp. Such microorganisms of oral microbiota as E. faecalis, Lactobacillus spp., Pseudomonas aeruginosa, and Candida spp. demonstrated statistically significant changes. All these changes indicate an improvement in the patients' condition in the experimental group compared to the control group. Our algorithm used for the development of personalized diets for patients with diabetes type 2 demonstrated clinical efficacy of its implementation.
Collapse
Affiliation(s)
- Tamara Meleshko
- Research Development and Educational Centre of Molecular Microbiology and Mucosal Immunology, Uzhhorod National University, Uzhhorod, Ukraine.,Department of Clinical Laboratory Diagnostics and Pharmacology, Faculty of Dentistry, Uzhhorod National University, Uzhhorod, Ukraine.,Ediens LLC, Uzhhorod, Ukraine
| | - Roman Rukavchuk
- Research Development and Educational Centre of Molecular Microbiology and Mucosal Immunology, Uzhhorod National University, Uzhhorod, Ukraine.
| | | | - Nadiya Boyko
- Research Development and Educational Centre of Molecular Microbiology and Mucosal Immunology, Uzhhorod National University, Uzhhorod, Ukraine.,Department of Clinical Laboratory Diagnostics and Pharmacology, Faculty of Dentistry, Uzhhorod National University, Uzhhorod, Ukraine.,Ediens LLC, Uzhhorod, Ukraine
| |
Collapse
|
682
|
Liu J, Liu F, Arıoğlu‐Tuncil S, Xie Z, Fu X, Huang Q, Zhang B. In vitro
faecal fermentation outcomes and microbiota shifts of resistant starch spherulites. Int J Food Sci Technol 2022. [DOI: 10.1111/ijfs.15237] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Jiaying Liu
- School of Food Science and Engineering Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety South China University of Technology Guangzhou 510640 China
| | - Feitong Liu
- H&H Group Global Research and Technology Center Guangzhou 510700 China
| | - Seda Arıoğlu‐Tuncil
- Department of Nutrition and Dietetics Faculty of Health Sciences Necmettin Erbakan University Konya 42090 Turkey
| | - Zhuqing Xie
- School of Food Science and Engineering Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety South China University of Technology Guangzhou 510640 China
- Department of Food Science University of Copenhagen Frederiksberg DK‐1958 Denmark
| | - Xiong Fu
- School of Food Science and Engineering Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety South China University of Technology Guangzhou 510640 China
- Overseas Expertise Introduction Center for Discipline Innovation of Food Nutrition and Human Health (111 Center) Guangzhou 510640 China
| | - Qiang Huang
- School of Food Science and Engineering Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety South China University of Technology Guangzhou 510640 China
- Overseas Expertise Introduction Center for Discipline Innovation of Food Nutrition and Human Health (111 Center) Guangzhou 510640 China
| | - Bin Zhang
- School of Food Science and Engineering Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety South China University of Technology Guangzhou 510640 China
- Overseas Expertise Introduction Center for Discipline Innovation of Food Nutrition and Human Health (111 Center) Guangzhou 510640 China
| |
Collapse
|
683
|
Du L, Li Q, Yi H, Kuang T, Tang Y, Fan G. Gut microbiota-derived metabolites as key actors in type 2 diabetes mellitus. Biomed Pharmacother 2022; 149:112839. [PMID: 35325852 DOI: 10.1016/j.biopha.2022.112839] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/14/2022] [Accepted: 03/16/2022] [Indexed: 12/01/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) is one of the most risk factors threatening human health. Although genetic and environmental factors contribute to the development of T2DM, gut microbiota has also been found to be involved. Gut microbiota-derived metabolites are a key factor in host-microbe crosstalk, and have been revealed to play a central role in the physiology and physiopathology of T2DM. In this review, we provide a timely and comprehensive summary of the microbial metabolites that are protective or causative for T2DM, including some amino acids-derived metabolites, short-chain fatty acids, trimethylamine N-oxide, and bile acids. The mechanisms by which metabolites affect T2DM have been elaborated. Knowing more about these processes will increase our understanding of the causal relationship between gut microbiota and T2DM. Moreover, some frontier therapies that target gut microbes and their metabolites to improve T2DM, including dietary intervention, fecal microbiota transplantation, probiotics, prebiotics or synbiotics intervention, and drugging microbial metabolism, have been critically discussed. This review may provide novel insights for the development of targeted and personalized treatments for T2DM based on gut microbial metabolites. More high-quality clinical trials are needed to accelerate the clinical translation of gut-targeted therapies for T2DM.
Collapse
Affiliation(s)
- Leilei Du
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Qi Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Huan Yi
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Tingting Kuang
- School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yi Tang
- Department of Endocrinology, Chengdu Fifth People's Hospital, Chengdu 611130, China.
| | - Gang Fan
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| |
Collapse
|
684
|
Xiao L, Zhang F, Zhao F. Large-scale microbiome data integration enables robust biomarker identification. NATURE COMPUTATIONAL SCIENCE 2022; 2:307-316. [PMID: 38177817 PMCID: PMC10766547 DOI: 10.1038/s43588-022-00247-8] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 04/12/2022] [Indexed: 01/06/2024]
Abstract
The close association between gut microbiota dysbiosis and human diseases is being increasingly recognized. However, contradictory results are frequently reported, as confounding effects exist. The lack of unbiased data integration methods is also impeding the discovery of disease-associated microbial biomarkers from different cohorts. Here we propose an algorithm, NetMoss, for assessing shifts of microbial network modules to identify robust biomarkers associated with various diseases. Compared to previous approaches, the NetMoss method shows better performance in removing batch effects. Through comprehensive evaluations on both simulated and real datasets, we demonstrate that NetMoss has great advantages in the identification of disease-related biomarkers. Based on analysis of pandisease microbiota studies, there is a high prevalence of multidisease-related bacteria in global populations. We believe that large-scale data integration will help in understanding the role of the microbiome from a more comprehensive perspective and that accurate biomarker identification will greatly promote microbiome-based medical diagnosis.
Collapse
Affiliation(s)
- Liwen Xiao
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Fengyi Zhang
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing, China
| | - Fangqing Zhao
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing, China.
- University of Chinese Academy of Sciences, Beijing, China.
- Key Laboratory of Systems Biology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China.
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
685
|
Anipindi M, Bitetto D. Diagnostic and Therapeutic Uses of the Microbiome in the Field of Oncology. Cureus 2022; 14:e24890. [PMID: 35698690 PMCID: PMC9184241 DOI: 10.7759/cureus.24890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/08/2022] [Indexed: 11/21/2022] Open
Abstract
Cancer is a leading cause of death worldwide and it can affect almost every part of the human body. Effective screening and early diagnosis of cancers is extremely difficult due to the multifactorial etiology of the disease and delayed presentation of the patients. The available treatments are usually not specific to the affected organ system, leading to intolerable systemic side effects and early withdrawal from therapies. In vivo and in vitro studies have revealed an association of specific microbiome signatures with individual cancers. The cancer-related human microbiome has also been shown to affect the response of tissues to chemotherapy, immunotherapy, and radiation. This is an excellent opportunity for us to design specific screening markers using the microbiome to prevent cancers and diagnose them early. We can also develop precise treatments that can target cancer-affected specific organ systems and probably use a lesser dose of chemotherapy or radiation for the same effect. This prevents adverse effects and early cessation of treatments. However, we need further studies to exactly clarify and characterize these associations. In this review article, we focus on the association of the microbiome with individual cancers and highlight its future role in cancer screenings, diagnosis, prognosis, and treatments.
Collapse
Affiliation(s)
- Manasa Anipindi
- Internal Medicine, Einstein Medical Center Philadelphia, East Norriton, USA
| | - Daniel Bitetto
- Internal Medicine, Einstein Medical Center Philadelphia, East Norriton, USA
| |
Collapse
|
686
|
Childhood body size directly increases type 1 diabetes risk based on a lifecourse Mendelian randomization approach. Nat Commun 2022; 13:2337. [PMID: 35484151 PMCID: PMC9051135 DOI: 10.1038/s41467-022-29932-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 04/08/2022] [Indexed: 12/14/2022] Open
Abstract
The rising prevalence of childhood obesity has been postulated as an explanation for the increasing rate of individuals diagnosed with type 1 diabetes (T1D). In this study, we use Mendelian randomization (MR) to provide evidence that childhood body size has an effect on T1D risk (OR = 2.05 per change in body size category, 95% CI = 1.20 to 3.50, P = 0.008), which remains after accounting for body size at birth and during adulthood using multivariable MR (OR = 2.32, 95% CI = 1.21 to 4.42, P = 0.013). We validate this direct effect of childhood body size using data from a large-scale T1D meta-analysis based on n = 15,573 cases and n = 158,408 controls (OR = 1.94, 95% CI = 1.21 to 3.12, P = 0.006). We also provide evidence that childhood body size influences risk of asthma, eczema and hypothyroidism, although multivariable MR suggested that these effects are mediated by body size in later life. Our findings support a causal role for higher childhood body size on risk of being diagnosed with T1D, whereas its influence on the other immune-associated diseases is likely explained by a long-term effect of remaining overweight for many years over the lifecourse. The rise in type 1 diabetes is thought to be related to increased childhood obesity, but this relationship is not well understood. In this study, the authors utilize Mendelian randomization to separate the direct and indirect effects of childhood body size on risk of type 1 diabetes and 7 other immune-associated disease outcomes.
Collapse
|
687
|
Puhlmann ML, Jokela R, van Dongen KCW, Bui TPN, van Hangelbroek RWJ, Smidt H, de Vos WM, Feskens EJM. Dried chicory root improves bowel function, benefits intestinal microbial trophic chains and increases faecal and circulating short chain fatty acids in subjects at risk for type 2 diabetes. GUT MICROBIOME (CAMBRIDGE, ENGLAND) 2022; 3:e4. [PMID: 39295776 PMCID: PMC11407914 DOI: 10.1017/gmb.2022.4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 02/02/2022] [Accepted: 04/12/2022] [Indexed: 09/21/2024]
Abstract
We investigated the impact of dried chicory root in a randomised, placebo-controlled trial with 55 subjects at risk for type 2 diabetes on bowel function, gut microbiota and its products, and glucose homeostasis. The treatment increased stool softness (+1.1 ± 0.3 units; p = 0.034) and frequency (+0.6 ± 0.2 defecations/day; p < 0.001), strongly modulated gut microbiota composition (7 % variation; p = 0.001), and dramatically increased relative levels (3-4-fold) of Anaerostipes and Bifidobacterium spp., in a dose-dependent, reversible manner. A synthetic community, including selected members of these genera and a Bacteroides strain, generated a butyrogenic trophic chain from the product. Faecal acetate, propionate and butyrate increased by 25.8 % (+13.0 ± 6.3 mmol/kg; p = 0.023) as did their fasting circulating levels by 15.7 % (+7.7 ± 3.9 μM; p = 0.057). In the treatment group the glycaemic coefficient of variation decreased from 21.3 ± 0.94 to 18.3 ± 0.84 % (p = 0.004), whereas fasting glucose and HOMA-ir decreased in subjects with low baseline Blautia levels (-0.3 ± 0.1 mmol/L fasting glucose; p = 0.0187; -0.14 ± 0.1 HOMA-ir; p = 0.045). Dried chicory root intake rapidly and reversibly affects bowel function, benefits butyrogenic trophic chains, and promotes glycaemic control.
Collapse
Affiliation(s)
- Marie-Luise Puhlmann
- Laboratory of Microbiology, Wageningen University & Research, Wageningen, The Netherlands
- Division of Human Nutrition and Health, Wageningen University & Research, Wageningen, The Netherlands
| | - Roosa Jokela
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Katja Catharina Wilhelmina van Dongen
- Laboratory of Microbiology, Wageningen University & Research, Wageningen, The Netherlands
- Division of Toxicology, Wageningen University & Research, Wageningen, The Netherlands
| | - Thi Phuong Nam Bui
- Laboratory of Microbiology, Wageningen University & Research, Wageningen, The Netherlands
- Caelus Health, Amsterdam, The Netherlands
| | - Roland Willem Jan van Hangelbroek
- Division of Human Nutrition and Health, Wageningen University & Research, Wageningen, The Netherlands
- Department of Data Science, Euretos BV, Utrecht, The Netherlands
| | - Hauke Smidt
- Laboratory of Microbiology, Wageningen University & Research, Wageningen, The Netherlands
| | - Willem Meindert de Vos
- Laboratory of Microbiology, Wageningen University & Research, Wageningen, The Netherlands
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | | |
Collapse
|
688
|
Yan L, Vaghari-Tabari M, Malakoti F, Moein S, Qujeq D, Yousefi B, Asemi Z. Quercetin: an effective polyphenol in alleviating diabetes and diabetic complications. Crit Rev Food Sci Nutr 2022; 63:9163-9186. [PMID: 35468007 DOI: 10.1080/10408398.2022.2067825] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Various studies, especially in recent years, have shown that quercetin has beneficial therapeutic effects in various human diseases, including diabetes. Quercetin has significant anti-diabetic effects and may be helpful in lowering blood sugar and increasing insulin sensitivity. Quercetin appears to affect many factors and signaling pathways involved in insulin resistance and the pathogenesis of type 2 of diabetes. TNFα, NFKB, AMPK, AKT, and NRF2 are among the factors that are affected by quercetin. In addition, quercetin can be effective in preventing and ameliorating the diabetic complications, including diabetic nephropathy, cardiovascular complications, neuropathy, delayed wound healing, and retinopathy, and affects the key mechanisms involved in the pathogenesis of these complications. These positive effects of quercetin may be related to its anti-inflammatory and anti-oxidant properties. In this article, after a brief review of the pathogenesis of insulin resistance and type 2 diabetes, we will review the latest findings on the anti-diabetic effects of quercetin with a molecular perspective. Then we will review the effects of quercetin on the key mechanisms of pathogenesis of diabetes complications including nephropathy, cardiovascular complications, neuropathy, delayed wound healing, and retinopathy. Finally, clinical trials investigating the effect of quercetin on diabetes and diabetes complications will be reviewed.
Collapse
Affiliation(s)
- Lei Yan
- Clinical Experimental Centre, Xi'an International Medical Center Hospital, Xi'an, China
- Department of Pre-Clinical Sciences, Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Kajang, Malaysia
| | - Mostafa Vaghari-Tabari
- Student's Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Faezeh Malakoti
- Student's Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Soheila Moein
- Medicinal Plants Processing Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Durdi Qujeq
- Cellular and Molecular Biology Research Center (CMBRC), Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Bahman Yousefi
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
689
|
Papadopoulos PD, Tsigalou C, Valsamaki PN, Konstantinidis TG, Voidarou C, Bezirtzoglou E. The Emerging Role of the Gut Microbiome in Cardiovascular Disease: Current Knowledge and Perspectives. Biomedicines 2022; 10:biomedicines10050948. [PMID: 35625685 PMCID: PMC9139035 DOI: 10.3390/biomedicines10050948] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 03/27/2022] [Accepted: 03/28/2022] [Indexed: 01/27/2023] Open
Abstract
The collection of normally non-pathogenic microorganisms that mainly inhabit our gut lumen shapes our health in many ways. Structural and functional perturbations in the gut microbial pool, known as “dysbiosis”, have been proven to play a vital role in the pathophysiology of several diseases, including cardiovascular disease (CVD). Although therapeutic regimes are available to treat this group of diseases, they have long been the main cause of mortality and morbidity worldwide. While age, sex, genetics, diet, tobacco use, and alcohol consumption are major contributors (World Health Organization, 2018), they cannot explain all of the consequences of CVD. In addition to the abovementioned traditional risk factors, the constant search for novel preventative and curative tools has shed light on the involvement of gut bacteria and their metabolites in the pathogenesis of CVD. In this narrative review, we will discuss the established interconnections between the gut microbiota and CVD, as well as the plausible therapeutic perspectives.
Collapse
Affiliation(s)
- Panagiotis D. Papadopoulos
- Master Programme Food, Nutrition and Microbiome, Department of Medicine, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (P.D.P.); (E.B.)
| | - Christina Tsigalou
- Master Programme Food, Nutrition and Microbiome, Department of Medicine, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (P.D.P.); (E.B.)
- Laboratory of Microbiology, Department of Medicine, Democritus University of Thrace, 68100 Alexandroupolis, Greece
- Correspondence:
| | - Pipitsa N. Valsamaki
- Nuclear Medicine Department, Medical School, Democritus University of Thrace, 68100 Alexandroupolis, Greece;
| | | | | | - Eugenia Bezirtzoglou
- Master Programme Food, Nutrition and Microbiome, Department of Medicine, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (P.D.P.); (E.B.)
- Laboratory of Hygiene and Environmental Protection, Department of Medicine, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| |
Collapse
|
690
|
Debédat J, Le Roy T, Voland L, Belda E, Alili R, Adriouch S, Bel Lassen P, Kasahara K, Hutchison E, Genser L, Torres L, Gamblin C, Rouault C, Zucker JD, Kapel N, Poitou C, Marcelin G, Rey FE, Aron-Wisnewsky J, Clément K. The human gut microbiota contributes to type-2 diabetes non-resolution 5-years after Roux-en-Y gastric bypass. Gut Microbes 2022; 14:2050635. [PMID: 35435140 PMCID: PMC9037437 DOI: 10.1080/19490976.2022.2050635] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Roux-en-Y gastric bypass (RYGB) is efficient at inducing drastic albeit variable weight loss and type-2 diabetes (T2D) improvements in patients with severe obesity and T2D. We hypothesized a causal implication of the gut microbiota (GM) in these metabolic benefits, as RYGB is known to deeply impact its composition. In a cohort of 100 patients with baseline T2D who underwent RYGB and were followed for 5-years, we used a hierarchical clustering approach to stratify subjects based on the severity of their T2D (Severe vs Mild) throughout the follow-up. We identified via nanopore-based GM sequencing that the more severe cases of unresolved T2D were associated with a major increase of the class Bacteroidia, including 12 species comprising Phocaeicola dorei, Bacteroides fragilis, and Bacteroides caecimuris. A key observation is that patients who underwent major metabolic improvements do not harbor this enrichment in Bacteroidia, as those who presented mild cases of T2D at all times. In a separate group of 36 patients with similar baseline clinical characteristics and preoperative GM sequencing, we showed that this increase in Bacteroidia was already present at baseline in the most severe cases of T2D. To explore the causal relationship linking this enrichment in Bacteroidia and metabolic alterations, we selected 13 patients across T2D severity clusters at 5-years and performed fecal matter transplants in mice. Our results show that 14 weeks after the transplantations, mice colonized with the GM of Severe donors have impaired glucose tolerance and insulin sensitivity as compared to Mild-recipients, all in the absence of any difference in body weight and composition. GM sequencing of the recipient animals revealed that the hallmark T2D-severity associated bacterial features were transferred and were associated with the animals' metabolic alterations. Therefore, our results further establish the GM as a key contributor to long-term glucose metabolism improvements (or lack thereof) after RYGB.
Collapse
Affiliation(s)
- Jean Debédat
- Nutrition and obesities; systemic approaches (NutriOmics), Sorbonne Université, INSERM, ParisFrance
| | - Tiphaine Le Roy
- Nutrition and obesities; systemic approaches (NutriOmics), Sorbonne Université, INSERM, ParisFrance
| | - Lise Voland
- Nutrition and obesities; systemic approaches (NutriOmics), Sorbonne Université, INSERM, ParisFrance
| | | | - Rohia Alili
- Nutrition and obesities; systemic approaches (NutriOmics), Sorbonne Université, INSERM, ParisFrance
| | - Solia Adriouch
- Nutrition and obesities; systemic approaches (NutriOmics), Sorbonne Université, INSERM, ParisFrance
| | - Pierre Bel Lassen
- Nutrition and obesities; systemic approaches (NutriOmics), Sorbonne Université, INSERM, ParisFrance,Assistance Publique Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Nutrition Department, France
| | - Kazuyuki Kasahara
- Department of Bacteriology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Evan Hutchison
- Department of Bacteriology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Laurent Genser
- Visceral Surgery Department, Assistance Publique Hôpitaux de Paris, Pitié-Salpêtrière Hospital, France
| | - Licia Torres
- Nutrition and obesities; systemic approaches (NutriOmics), Sorbonne Université, INSERM, ParisFrance
| | - Camille Gamblin
- Nutrition and obesities; systemic approaches (NutriOmics), Sorbonne Université, INSERM, ParisFrance
| | - Christine Rouault
- Nutrition and obesities; systemic approaches (NutriOmics), Sorbonne Université, INSERM, ParisFrance
| | - Jean-Daniel Zucker
- Nutrition and obesities; systemic approaches (NutriOmics), Sorbonne Université, INSERM, ParisFrance,Unité de Modélisation Mathématique et Informatique des Systèmes Complexes, UMMISCO, Sorbonne Universités, Institut de Recherche pour le Développement (IRD), France
| | - Nathalie Kapel
- Functional Coprology Department, Assistance Publique Hôpitaux de Paris, Pitié-Salpêtrière Hospital, France
| | - Christine Poitou
- Nutrition and obesities; systemic approaches (NutriOmics), Sorbonne Université, INSERM, ParisFrance,Assistance Publique Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Nutrition Department, France
| | - Geneviève Marcelin
- Nutrition and obesities; systemic approaches (NutriOmics), Sorbonne Université, INSERM, ParisFrance
| | - Federico E. Rey
- Department of Bacteriology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Judith Aron-Wisnewsky
- Nutrition and obesities; systemic approaches (NutriOmics), Sorbonne Université, INSERM, ParisFrance,Assistance Publique Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Nutrition Department, France,CONTACT Judith Aron-Wisnewsky Sorbonne Université, INSERM, Nutrition and obesities; systemic approaches (NutriOmics), Nutrition and obesities; systemic approaches (NutriOmics)75013, Paris, France
| | - Karine Clément
- Nutrition and obesities; systemic approaches (NutriOmics), Sorbonne Université, INSERM, ParisFrance,Assistance Publique Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Nutrition Department, France,Karine Clément Nutrition and obesities; systemic approaches (NutriOmics) Assistance Publique Hôpitaux de Paris, Pitié-Salpêtrière Hospital, ParisFranceNutrition Department
| |
Collapse
|
691
|
Bongers KS, McDonald RA, Winner KM, Falkowski NR, Brown CA, Baker JM, Hinkle KJ, Fergle DJ, Dickson RP. Antibiotics cause metabolic changes in mice primarily through microbiome modulation rather than behavioral changes. PLoS One 2022; 17:e0265023. [PMID: 35298489 PMCID: PMC8929607 DOI: 10.1371/journal.pone.0265023] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 02/20/2022] [Indexed: 12/15/2022] Open
Abstract
Background The microbiome is an important and increasingly-studied mediator of organismal metabolism, although how the microbiome affects metabolism remains incompletely understood. Many investigators use antibiotics to experimentally perturb the microbiome. However, antibiotics have poorly understood yet profound off-target effects on behavior and diet, including food and water aversion, that can confound experiments and limit their applicability. We thus sought to determine the relative influence of microbiome modulation and off-target antibiotic effects on the behavior and metabolic activity of mice. Results Mice treated with oral antibiotics via drinking water exhibited significant weight loss in fat, liver, and muscle tissue. These mice also exhibited a reduction in water and food consumption, with marked variability across antibiotic regimens. While administration of bitter-tasting but antimicrobially-inert compounds caused a similar reduction in water consumption, this did not cause tissue weight loss or reduced food consumption. Mice administered intraperitoneal antibiotics (bypassing the gastrointestinal tract) exhibited reduced tissue weights and oral intake, comparable to the effects of oral antibiotics. Antibiotic-treated germ-free mice did not have reduced tissue weights, providing further evidence that direct microbiome modulation (rather than behavioral effects) mediates these metabolic changes. Conclusions While oral antibiotics cause profound effects on food and water consumption, antibiotic effects on organismal metabolism are primarily mediated by microbiome modulation. We demonstrate that tissue-specific weight loss following antibiotic administration is due primarily to microbiome effects rather than food and water aversion, and identify antibiotic regimens that effectively modulate gut microbiota while minimizing off-target behavioral effects.
Collapse
Affiliation(s)
- Kale S. Bongers
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Health System, Ann Arbor, Michigan, United States of America
| | - Roderick A. McDonald
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Health System, Ann Arbor, Michigan, United States of America
| | - Katherine M. Winner
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Health System, Ann Arbor, Michigan, United States of America
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Nicole R. Falkowski
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Health System, Ann Arbor, Michigan, United States of America
| | - Christopher A. Brown
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Health System, Ann Arbor, Michigan, United States of America
- Institute for Research on Innovation and Science, Institute for Social Research, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Jennifer M. Baker
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Health System, Ann Arbor, Michigan, United States of America
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Kevin J. Hinkle
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Health System, Ann Arbor, Michigan, United States of America
| | - Daniel J. Fergle
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Health System, Ann Arbor, Michigan, United States of America
| | - Robert P. Dickson
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Health System, Ann Arbor, Michigan, United States of America
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
- Weil Institute for Critical Care Research and Innovation, Ann Arbor, Michigan, United States of America
- * E-mail:
| |
Collapse
|
692
|
Chen YZ, Gu J, Chuang WT, Du YF, Zhang L, Lu ML, Xu JY, Li HQ, Liu Y, Feng HT, Li YH, Qin LQ. Slowly Digestible Carbohydrate Diet Ameliorates Hyperglycemia and Hyperlipidemia in High-Fat Diet/Streptozocin-Induced Diabetic Mice. Front Nutr 2022; 9:854725. [PMID: 35495933 PMCID: PMC9051025 DOI: 10.3389/fnut.2022.854725] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 03/22/2022] [Indexed: 12/24/2022] Open
Abstract
Objective Given that the prevalence rate of type 2 diabetes mellitus (T2DM) continues to increase, it is important to find an effective method to prevent or treat this disease. Previous studies have shown that dietary intervention with a slowly digestible carbohydrate (SDC) diet can improve T2DM with almost no side effects. However, the underlying mechanisms of SDC protect against T2DM remains to be elucidated. Methods The T2DM mice model was established with a high-fat diet and streptozocin injection. Then, SDC was administered for 6 weeks. Bodyweight, food intake, organ indices, fasting blood glucose (FBG), oral glucose tolerance test (OGTT), homeostasis model assessment for insulin resistance (HOMA-IR), and other biochemical parameters were measured. Histopathological and lipid accumulation analyses were performed, and the glucose metabolism-related gene expressions in the liver and skeletal muscle were determined. Lastly, colonic microbiota was also analyzed. Results SDC intervention alleviated the weight loss in the pancreas, lowered blood glucose and glycosylated hemoglobin levels, and improved glucose tolerance and HOMA-IR. SDC intervention improved serum lipid profile, adipocytokines levels, and lowered the lipid accumulation in the liver, subcutaneous adipose tissue, and epididymal visceral adipose tissue. In addition, SDC intervention increased the expression levels of IRS-2 and GLUT-2 in liver tissues and elevated GLUT-4 expression levels in skeletal muscle tissues. Notably, SDC intervention decreased the Bacteroidetes/Firmicutes ratio, increased Desulfovibrio and Lachnospiraceae genus levels, and inhibited the relative abundance of potentially pathogenic bacteria. Conclusions SDC intervention can improve hyperglycemia and hyperlipidemia status in diabetic mice, suggesting that this intervention might be beneficial for T2DM.
Collapse
Affiliation(s)
- Yu-Zhong Chen
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Soochow University, Suzhou, China
| | - Jia Gu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Soochow University, Suzhou, China
| | - Wei-Ting Chuang
- Inner Mongolia Dairy Technology Research Institute Co., Ltd., Hohhot, China
| | - Ya-Fang Du
- Department of Nutrition and Food Hygiene, School of Public Health, Soochow University, Suzhou, China
| | - Lin Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, Soochow University, Suzhou, China
| | - Meng-Lan Lu
- Department of Nutrition and Food Hygiene, School of Public Health, Soochow University, Suzhou, China
| | - Jia-Ying Xu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Soochow University, Suzhou, China
| | - Hao-Qiu Li
- Inner Mongolia Dairy Technology Research Institute Co., Ltd., Hohhot, China
| | - Yan Liu
- Inner Mongolia Dairy Technology Research Institute Co., Ltd., Hohhot, China
| | - Hao-Tian Feng
- Inner Mongolia Dairy Technology Research Institute Co., Ltd., Hohhot, China
- *Correspondence: Hao-Tian Feng
| | - Yun-Hong Li
- Department of Nutrition and Food Hygiene, School of Public Health, Soochow University, Suzhou, China
- Yun-Hong Li
| | - Li-Qiang Qin
- Department of Nutrition and Food Hygiene, School of Public Health, Soochow University, Suzhou, China
- Li-Qiang Qin
| |
Collapse
|
693
|
Sevcuka A, White K, Terry C. Factors That Contribute to hIAPP Amyloidosis in Type 2 Diabetes Mellitus. Life (Basel) 2022; 12:life12040583. [PMID: 35455074 PMCID: PMC9025880 DOI: 10.3390/life12040583] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 04/01/2022] [Accepted: 04/12/2022] [Indexed: 12/24/2022] Open
Abstract
Cases of Type 2 Diabetes Mellitus (T2DM) are increasing at an alarming rate due to the rise in obesity, sedentary lifestyles, glucose-rich diets and other factors. Numerous studies have increasingly illustrated the pivotal role that human islet amyloid polypeptide (hIAPP) plays in the pathology of T2DM through damage and subsequent loss of pancreatic β-cell mass. HIAPP can misfold and form amyloid fibrils which are preceded by pre-fibrillar oligomers and monomers, all of which have been linked, to a certain extent, to β-cell cytotoxicity through a range of proposed mechanisms. This review provides an up-to-date summary of recent progress in the field, highlighting factors that contribute to hIAPP misfolding and aggregation such as hIAPP protein concentration, cell stress, molecular chaperones, the immune system response and cross-seeding with other amyloidogenic proteins. Understanding the structure of hIAPP and how these factors affect amyloid formation will help us better understand how hIAPP misfolds and aggregates and, importantly, help identify potential therapeutic targets for inhibiting amyloidosis so alternate and more effective treatments for T2DM can be developed.
Collapse
|
694
|
Guo Y, Zhu G, Wang F, Zhang H, Chen X, Mao Y, Lv Y, Xia F, Jin Y, Ding G, Yu J. Distinct Serum and Fecal Metabolite Profiles Linking With Gut Microbiome in Older Adults With Frailty. Front Med (Lausanne) 2022; 9:827174. [PMID: 35479954 PMCID: PMC9035822 DOI: 10.3389/fmed.2022.827174] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 03/10/2022] [Indexed: 12/16/2022] Open
Abstract
Frailty is a critical aging-related syndrome but the underlying metabolic mechanism remains poorly understood. The aim of this study was to identify novel biomarkers and reveal potential mechanisms of frailty based on the integrated analysis of metabolome and gut microbiome. In this study, twenty subjects consisted of five middle-aged adults and fifteen older adults, of which fifteen older subjects were divided into three groups: non-frail, pre-frail, and frail, with five subjects in each group. The presence of frailty, pre-frailty, or non-frailty was established according to the physical frailty phenotype (PFP). We applied non-targeted metabolomics to serum and feces samples and used 16S rDNA gene sequencing to detect the fecal microbiome. The associations between metabolites and gut microbiota were analyzed by the Spearman’s correlation analysis. Serum metabolic shifts in frailty mainly included fatty acids and derivatives, carbohydrates, and monosaccharides. Most of the metabolites belonging to these classes increased in the serum of frail older adults. Propylparaben was found to gradually decrease in non-frail, pre-frail, and frail older adults. Distinct changes in fecal metabolite profiles and gut microbiota were also found among middle-aged adults, non-frail and frail older subjects. The relative abundance of Faecalibacteriu, Roseburia, and Fusicatenibacter decreased while the abundance of Parabacteroides and Bacteroides increased in frailty. The above altered microbes were associated with the changed serum metabolites in frailty, which included dodecanedioic acid, D-ribose, D-(-)-mannitol, creatine and indole, and their related fecal metabolites. The changed microbiome and related metabolites may be used as the biomarkers of frailty and is worthy of further mechanistic studies.
Collapse
Affiliation(s)
- Yan Guo
- Division of Geriatric Endocrinology, Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Department of Neurology, Yancheng City No. 1 People’s Hospital, Yancheng, China
| | - Guoqin Zhu
- Division of Geriatric Gastroenterology, Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Fengliang Wang
- Department of Breast Surgery, Nanjing Maternity and Child Health Care Hospital, The Affiliated Obstetrics and Gynaecology Hospital of Nanjing Medical University, Nanjing, China
| | - Haoyu Zhang
- Division of Geriatric Endocrinology, Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Department of Human Biology Undergraduate, University of Toronto, Toronto, ON, Canada
| | - Xin Chen
- Division of Geriatric Endocrinology, Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yan Mao
- Division of Geriatric Endocrinology, Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yifan Lv
- Division of Geriatric Endocrinology, Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Fan Xia
- Division of Geriatric Endocrinology, Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yi Jin
- Division of Geriatric Endocrinology, Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Guoxian Ding
- Division of Geriatric Endocrinology, Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Guoxian Ding,
| | - Jing Yu
- Division of Geriatric Endocrinology, Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- *Correspondence: Jing Yu,
| |
Collapse
|
695
|
Species-level gut microbiota analysis in ovariectomized osteoporotic rats by Shallow shotgun sequencing. Gene 2022; 817:146205. [PMID: 35063575 DOI: 10.1016/j.gene.2022.146205] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 11/27/2021] [Accepted: 01/13/2022] [Indexed: 12/16/2022]
Abstract
Gut microbiota was verified to regulate bone metabolism and was closely associated with osteoporosis. Using 16S rRNA sequencing, gut microbiota at genus level such as Helicobacter, Bacteroides, and Prevotella were found to increase in the osteoporotic animals and people. However, the changes of species-level gut microbiota and related functional alterations were still unknown. Female SD rats were divided into the ovariectomized (OVX) group and the control group, and the fecal samples were collected at 4, 8, and 12 weeks to analyze the information of gut microbiota. Using Shallow shotgun sequencing, we compared the species-level gut microbiota structure, composition, and functional pathways of the OVX group with the control group. Alpha diversity of the OVX rats were significantly decreased than those in the control group. Beta diversity showed that samples in the two groups could be distinguished in each coordinate at different time points. Furthermore, the relative abundance of gut microbiota at species-level and differential analysis found that bacteria species such as Helicobacter rodentium, Lachnospiraceae bacterium 10 1, and Lachnospiraceae bacterium A4 were markedly increased in the OVX rats. Furthermore, differential analysis of KEGG functional pathway revealed that lysine metabolism was enriched in the OVX group.In conclusion, gut microbiota were significantly altered in structure and composition estrogen-deficiency osteoporotic rats at the species level. Functional metabolism of gut microbiota was also changed in osteoporotic group. These changes in gut microbiota at the species level might be closely associated with osteoporosis caused by estrogen deficiency.
Collapse
|
696
|
Chow E, Yang A, Chung CHL, Chan JCN. A Clinical Perspective of the Multifaceted Mechanism of Metformin in Diabetes, Infections, Cognitive Dysfunction, and Cancer. Pharmaceuticals (Basel) 2022; 15:ph15040442. [PMID: 35455439 PMCID: PMC9030054 DOI: 10.3390/ph15040442] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 03/30/2022] [Accepted: 03/30/2022] [Indexed: 12/16/2022] Open
Abstract
In type 2 diabetes, ecological and lifecourse factors may interact with the host microbiota to influence expression of his/her genomes causing perturbation of interconnecting biological pathways with diverse clinical course. Metformin is a plant-based or plant-derived medicinal product used for the treatment of type 2 diabetes for over 60 years and is an essential drug listed by the World Health Organization. By reducing mitochondrial oxidative phosphorylation and adenosine triphosphate (ATP) production, metformin increased AMP (adenosine monophosphate)-activated protein kinase (AMPK) activity and altered cellular redox state with reduced glucagon activity, endogenous glucose production, lipogenesis, and protein synthesis. Metformin modulated immune response by directly reducing neutrophil to lymphocyte ratio and improving the phagocytic function of immune cells. By increasing the relative abundance of mucin-producing and short-chain-fatty-acid-producing gut microbes, metformin further improved the host inflammatory and metabolic milieu. Experimentally, metformin promoted apoptosis and reduced proliferation of cancer cells by reducing their oxygen consumption and modulating the microenvironment. Both clinical and mechanistic studies support the pluripotent effects of metformin on reducing cardiovascular–renal events, infection, cancer, cognitive dysfunction, and all-cause death in type 2 diabetes, making this low-cost medication a fundamental therapy for individualization of other glucose-lowering drugs in type 2 diabetes. Further research into the effects of metformin on cognitive function, infection and cancer, especially in people without diabetes, will provide new insights into the therapeutic value of metformin in our pursuit of prevention and treatment of ageing-related as well as acute and chronic diseases beyond diabetes.
Collapse
Affiliation(s)
- Elaine Chow
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong 999077, China; (E.C.); (A.Y.); (C.H.L.C.)
- The Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong 999077, China
- Phase 1 Clinical Trial Centre, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong 999077, China
| | - Aimin Yang
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong 999077, China; (E.C.); (A.Y.); (C.H.L.C.)
- The Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong 999077, China
| | - Colin H. L. Chung
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong 999077, China; (E.C.); (A.Y.); (C.H.L.C.)
| | - Juliana C. N. Chan
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong 999077, China; (E.C.); (A.Y.); (C.H.L.C.)
- The Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong 999077, China
- Correspondence: ; Tel.: +852-3505-3138
| |
Collapse
|
697
|
Silva JSC, Seguro CS, Naves MMV. Gut microbiota and physical exercise in obesity and diabetes - A systematic review. Nutr Metab Cardiovasc Dis 2022; 32:863-877. [PMID: 35227549 DOI: 10.1016/j.numecd.2022.01.023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 01/14/2022] [Accepted: 01/17/2022] [Indexed: 11/20/2022]
Abstract
BACKGROUND AND AIM The gut microbiota (GM) plays an essential role in maintaining health, and imbalance in its composition is associated with the physiopathogenesis of metabolic diseases, such as obesity and type 2 diabetes mellitus (T2DM). Diet and antibiotics are known modulators of GM, but the influence of physical exercise in modulating the diversity and abundance of hindgut bacteria is still poorly understood. The aim of this systematic review was to investigate the scientific evidence about the effect of physical exercise on GM modulation in subjects with obesity and T2DM. METHODS AND RESULTS A search in PubMed, Web of Science, Scopus, Cochrane and Embase databases using keywords related to gut microbiota, physical exercise and metabolic diseases was performed. Eight clinical studies met the inclusion criteria, six in subjects with obesity and two in individuals with T2DM. In three studies carried out in individuals with obesity, exercise was able to positively modulate the diversity of GM and the abundance of some species of bacteria, mostly by increasing the Bifidobacteriaceae family, and the Bacteroides and Akkermansia genera, and by decreasing the Proteobacteria phylum. The studies in subjects with T2DM found that physical exercise may reduce metabolic endotoxemia markers. CONCLUSIONS Physical exercise may be a beneficial modulation strategy of GM composition in metabolic diseases, specifically aerobic exercises carried out for at least 6 weeks with moderate or high intensity. Nevertheless, well-designed clinical trials are needed to clarify the role of physical exercise on GM in subjects with obesity and T2DM.
Collapse
Affiliation(s)
- John S C Silva
- Scholl of Nutrition, Federal University of Goiás, Goiânia, GO, Brazil
| | - Camila S Seguro
- Scholl of Nutrition, Federal University of Goiás, Goiânia, GO, Brazil
| | | |
Collapse
|
698
|
Michels N, Zouiouich S, Vanderbauwhede B, Vanacker J, Indave Ruiz BI, Huybrechts I. Human microbiome and metabolic health: An overview of systematic reviews. Obes Rev 2022; 23:e13409. [PMID: 34978141 DOI: 10.1111/obr.13409] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 10/05/2021] [Accepted: 11/28/2021] [Indexed: 12/16/2022]
Abstract
To summarize the microbiome's role in metabolic disorders (insulin resistance, hyperglycemia, type 2 diabetes, obesity, hyperlipidemia, hypertension, nonalcoholic fatty liver disease [NAFLD], and metabolic syndrome), systematic reviews on observational or interventional studies (prebiotics/probiotics/synbiotics/transplant) were searched in MEDLINE and Embase until September 2020. The 87 selected systematic reviews included 57 meta-analyses. Methodological quality (AMSTAR2) was moderate in 62%, 12% low, and 26% critically low. Observational studies on obesity (10 reviews) reported less gut bacterial diversity with higher Fusobacterium, Lactobacillus reuteri, Bacteroides fragilis, and Staphylococcus aureus, whereas lower Methanobrevibacter, Lactobacillus plantarum, Akkermansia muciniphila, and Bifidobacterium animalis compared with nonobese. For diabetes (n = 1), the same was found for Fusobacterium and A. muciniphila, whereas higher Ruminococcus and lower Faecalibacterium, Roseburia, Bacteroides vulgatus, and several Bifidobacterium spp. For NAFLD (n = 2), lower Firmicutes, Rikenellaceae, Ruminococcaceae, whereas higher Escherichia and Lactobacillus were detected. Discriminating bacteria overlapped between metabolic disorders, those with high abundance being often involved in inflammation, whereas those with low abundance being used as probiotics. Meta-analyses (n = 54) on interventional studies reported 522 associations: 54% was statistically significant with intermediate effect size and moderate between-study heterogeneity. Meta-evidence was highest for probiotics and lowest for fecal transplant. Future avenues include better methodological quality/comparability, testing functional differences, new intervention strategies, and considerating other body habitats and kingdoms.
Collapse
Affiliation(s)
- Nathalie Michels
- Department of Public Health and Primary Care, Ghent University, Ghent, Belgium
| | - Semi Zouiouich
- International Agency for Research on Cancer, Lyon, France
| | - Bert Vanderbauwhede
- Department of Public Health and Primary Care, Ghent University, Ghent, Belgium
| | - Judith Vanacker
- Department of Public Health and Primary Care, Ghent University, Ghent, Belgium
| | | | | |
Collapse
|
699
|
Maan HS, Chaurasia D, Kapoor G, Dave L, Siddiqui A, Pal S, Singh HO, Biswas D, Chowdhary R. Intestinal viral infections of nSARS-CoV2 in the Indian community: Risk of virus spread in India. J Med Virol 2022; 94:1315-1329. [PMID: 34825708 PMCID: PMC9015588 DOI: 10.1002/jmv.27480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Revised: 11/14/2021] [Accepted: 11/24/2021] [Indexed: 12/15/2022]
Abstract
In December 2019, novel severe acute respiratory syndrome coronavirus 2 (nSARS-CoV-2) virus outbreaks emerged from Wuhan, China, and spread all over the world, including India. Molecular diagnosis of Coronavirus Disease 2019 (COVID) 19 for densely and highly populated countries like India is time-consuming. A few reports have described the successful diagnosis of nSARS-CoV-2 virus from sewage and wastewater samples contaminated with fecal matter, suggesting the diagnosis of COVID 19 from the same to raise an alarm about the community transmission of virus for implementation of evacuation and lockdown strategies. So far, the association between the detection of virus and its concentration in stool samples with severity of the disease and the presence or absence of gastrointestinal symptoms have been rarely reported. We led the search utilizing multiple databases, specifically PubMed (Medline), EMBASE, and Google Scholar. We conducted a literature survey on gastrointestinal infection and the spread of this virus through fecal-oral transmission. Reports suggested that the existence and persistence of nSARS-CoV-2 in anal/rectal swabs and stool specimens for a longer period of time than in nasopharyngeal swabs provides a strong tenable outcome of gastrointestinal contamination and dissemination of this infection via potential fecal-oral transmission. This review may be helpful to conduct further studies to address the enteric involvement and excretion of nSARS-CoV-2 RNA in feces and control the community spread in both COVID-19 patients ahead of the onset of symptoms and in asymptomatic individuals through wastewater and sewage surveillance as an early indication of infection. The existence of the viral genome and active viral particle actively participate in genomic variations. Hence, we comprehended the enteric spread of different viruses amongst communities with special reference to nSARS-CoV-2.
Collapse
Affiliation(s)
- Harjeet S. Maan
- State Virology Laboratory, Department of MicrobiologyGandhi Medical CollegeBhopalMadhya PradeshIndia
| | - Deepti Chaurasia
- Department of MicrobiologyGandhi Medical CollegeBhopalMadhya PradeshIndia
| | - Garima Kapoor
- Department of MicrobiologyGandhi Medical CollegeBhopalMadhya PradeshIndia
| | - Lokendra Dave
- Department of Respiratory MedicineGandhi Medical CollegeBhopalMadhya PradeshIndia
| | - Arshi Siddiqui
- Department of BiotechnologyBarkatullah UniversityBhopalMadhya PradeshIndia
| | - Savita Pal
- Department of BiochemistryCentral Drug Research InstituteLucknowUttar PradeshIndia
| | - Hari O. Singh
- Division of Molecular Biology, Indian Council of Medical ResearchNational AIDS Research InstitutePuneMaharashtraIndia
| | - Debasis Biswas
- Department of MicrobiologyAll India Institute of Medical Sciences BhopalBhopalMadhya PradeshIndia
| | - Rashmi Chowdhary
- Department of BiochemistryAll India Institute of Medical Sciences BhopalBhopalMadhya PradeshIndia
| |
Collapse
|
700
|
Vijay A, Valdes AM. Role of the gut microbiome in chronic diseases: a narrative review. Eur J Clin Nutr 2022; 76:489-501. [PMID: 34584224 PMCID: PMC8477631 DOI: 10.1038/s41430-021-00991-6] [Citation(s) in RCA: 219] [Impact Index Per Article: 73.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 06/29/2021] [Accepted: 07/27/2021] [Indexed: 02/07/2023]
Affiliation(s)
- Amrita Vijay
- Division of Rheumatology, Orthopaedics and Dermatology, School of Medicine, The University of Nottingham, Nottingham, UK.
| | - Ana M Valdes
- Division of Rheumatology, Orthopaedics and Dermatology, School of Medicine, The University of Nottingham, Nottingham, UK
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and University of Nottingham, Nottingham, UK
| |
Collapse
|