1
|
Zhang T, Liu J, Liu X, Wang Q, Zhang H. The causal impact of gut microbiota on circulating adipokine concentrations: a two-sample Mendelian randomization study. Hormones (Athens) 2024; 23:789-799. [PMID: 38564143 DOI: 10.1007/s42000-024-00553-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 03/19/2024] [Indexed: 04/04/2024]
Abstract
PURPOSE Evidence from previous experimental and observational research demonstrates that the gut microbiota is related to circulating adipokine concentrations. Nevertheless, the debate as to whether gut microbiome composition causally influences circulating adipokine concentrations remains unresolved. This study aimed to take an essential step in elucidating this issue. METHODS We used two-sample Mendelian randomization (MR) to causally analyze genetic variation statistics for gut microbiota and four adipokines (including adiponectin, leptin, soluble leptin receptor [sOB-R], and plasminogen activator inhibitor-1 [PAI-1]) from large-scale genome-wide association studies (GWAS) datasets. A range of sensitivity analyses was also conducted to assess the stability and reliability of the results. RESULTS The composite results of the MR and sensitivity analyses revealed 22 significant causal associations. In particular, there is a suggestive causality between the family Clostridiaceae1 (IVW: β = 0.063, P = 0.034), the genus Butyrivibrio (IVW: β = 0.029, P = 0.031), and the family Alcaligenaceae (IVW: β=-0.070, P = 0.014) and adiponectin. Stronger causal effects with leptin were found for the genus Enterorhabdus (IVW: β=-0.073, P = 0.038) and the genus Lachnospiraceae (NK4A136 group) (IVW: β=-0.076, P = 0.01). Eight candidate bacterial groups were found to be associated with sOB-R, with the phylum Firmicutes (IVW: β = 0.235, P = 0.03) and the order Clostridiales (IVW: β = 0.267, P = 0.028) being of more interest. In addition, the genus Roseburia (IVW: β = 0.953, P = 0.022) and the order Lactobacillales (IVW: β=-0.806, P = 0.042) were suggestive of an association with PAI-1. CONCLUSION This study reveals a causal relationship between the gut microbiota and circulating adipokines and may help to offer novel insights into the prevention of abnormal concentrations of circulating adipokines and obesity-related diseases.
Collapse
Affiliation(s)
- Tongxin Zhang
- Department of Ultrasound, Shandong Provincial Hospital, Shandong First Medical University, Jinan, Shandong, China
| | - Jingyu Liu
- Department of Ultrasound, Shandong Provincial Hospital, Shandong First Medical University, Jinan, Shandong, China
| | - Xiao Liu
- Department of Ultrasound, Shandong Provincial Hospital, Shandong First Medical University, Jinan, Shandong, China
| | - Qian Wang
- Department of Ultrasound, Shandong Provincial Hospital, Shandong First Medical University, Jinan, Shandong, China.
| | - Huawei Zhang
- Department of Ultrasound, Shandong Provincial Hospital, Shandong First Medical University, Jinan, Shandong, China.
| |
Collapse
|
2
|
HUANG X, LI Y, ZHU C, ZHU H, JIANG C, ZHU X, ZHANG C, JIN C. Weitiao No. 3 (3) enhances the efficacy of anti-programmed cell death protein-1 immunotherapy by modulating the intestinal microbiota in an orthotopic model of gastric cancer mice. J TRADIT CHIN MED 2024; 44:906-915. [PMID: 39380221 PMCID: PMC11462543 DOI: 10.19852/j.cnki.jtcm.2024.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Accepted: 12/05/2023] [Indexed: 10/10/2024]
Abstract
OBJECTIVE To explore the effects of Weitiao No. 3 (3, WD-3) on anti-programmed cell death protein-1 (PD-1) immunotherapy in gastric cancer (GC). METHODS The intestinal microbiota was analyzed by 16S rDNA sequencing of fecal samples from three groups: healthy people (Health), GC patients (GC), and WD-3-treated GC patients (WD-3). Next, we established an orthotopic model of GC mice, which were treated with anti-PD-1, WD-3, or an inoculation of intestinal bacteria. Immune markers CD3, CD4, CD8, and forkhead box protein P3 (FOXP3), and the cell proliferation marker Ki67, were evaluated by immunohistochemistry. Cell apoptosis in GC tumors was assessed by terminal-deoxynucleotidyl-transferase-mediated deoxyuridine triphosphate nick end labeling staining. Enzyme-linked immunosorbent assays (ELISAs) were performed to analyze the serum levels of the following cytokines in GC mice: tumor necrosis factor (TNF)-α, interleukin (IL)-2, IL-6, IL-10, interferon (IFN)-γ, and transforming growth factor (TGF)-β. RESULTS Sequencing data showed that there were significant differences in the composition of the gut microbial community among the three human groups. The gut bacteria in the three groups mainly comprised the phyla Firmicutes, Proteobacteria, Bacteroidetes, and Actinobacteria. At the genus level, the relative abundances of Bifidobacterium and Coprococcus showed significant decreases in the GC group, and an obvious increase in the WD-3 group, compared with the Health group. Interestingly, the relative abundance of Saccharopolyspora was only detected in the WD-3 group. The results of in vivo experiments in GC mice showed that WD-3 or anti-PD-1 treatment increased the levels of CD3+, CD4+, and CD8+ T cells, but decreased the levels of FOXP3+ regulatory T cells. Furthermore, WD-3 or PD-1 antibody treatment inhibited proliferation and promoted apoptosis of GC tumor cells. ELISA analysis showed that WD-3 or PD-1 antibody treatment facilitated TNF-α, IL-2, and IFN-γ expression, while suppressing IL-6, IL-10, and TGF-β expression. Combination therapy with WD-3 and anti-PD-1 intensified all of these effects. CONCLUSION WD-3 enhanced the immunotherapeutic efficacy of anti-PD-1 by modulating the intestinal microbiota in an orthotopic model of GC mice.
Collapse
Affiliation(s)
- Xiaona HUANG
- Department of Oncology, Wuxi Hospital Affiliated to Nanjing University of Chinese Medicine (Wuxi Hospital of Traditional Chinese Medicine), Wuxi 214071, China
| | - Yuzhen LI
- Department of Oncology, Wuxi Hospital Affiliated to Nanjing University of Chinese Medicine (Wuxi Hospital of Traditional Chinese Medicine), Wuxi 214071, China
| | - Chenyang ZHU
- Department of Oncology, Wuxi Hospital Affiliated to Nanjing University of Chinese Medicine (Wuxi Hospital of Traditional Chinese Medicine), Wuxi 214071, China
| | - Hengzhou ZHU
- Department of Oncology, Wuxi Hospital Affiliated to Nanjing University of Chinese Medicine (Wuxi Hospital of Traditional Chinese Medicine), Wuxi 214071, China
| | - Chenyu JIANG
- Department of Oncology, Wuxi Hospital Affiliated to Nanjing University of Chinese Medicine (Wuxi Hospital of Traditional Chinese Medicine), Wuxi 214071, China
| | - Xiaodan ZHU
- Department of Oncology, Wuxi Hospital Affiliated to Nanjing University of Chinese Medicine (Wuxi Hospital of Traditional Chinese Medicine), Wuxi 214071, China
| | - Chencen ZHANG
- Department of Oncology, Wuxi Hospital Affiliated to Nanjing University of Chinese Medicine (Wuxi Hospital of Traditional Chinese Medicine), Wuxi 214071, China
| | - Chunhui JIN
- Department of Oncology, Wuxi Hospital Affiliated to Nanjing University of Chinese Medicine (Wuxi Hospital of Traditional Chinese Medicine), Wuxi 214071, China
| |
Collapse
|
3
|
Fan L, Xia Y, Wang Y, Han D, Liu Y, Li J, Fu J, Wang L, Gan Z, Liu B, Fu J, Zhu C, Wu Z, Zhao J, Han H, Wu H, He Y, Tang Y, Zhang Q, Wang Y, Zhang F, Zong X, Yin J, Zhou X, Yang X, Wang J, Yin Y, Ren W. Gut microbiota bridges dietary nutrients and host immunity. SCIENCE CHINA. LIFE SCIENCES 2023; 66:2466-2514. [PMID: 37286860 PMCID: PMC10247344 DOI: 10.1007/s11427-023-2346-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Accepted: 04/05/2023] [Indexed: 06/09/2023]
Abstract
Dietary nutrients and the gut microbiota are increasingly recognized to cross-regulate and entrain each other, and thus affect host health and immune-mediated diseases. Here, we systematically review the current understanding linking dietary nutrients to gut microbiota-host immune interactions, emphasizing how this axis might influence host immunity in health and diseases. Of relevance, we highlight that the implications of gut microbiota-targeted dietary intervention could be harnessed in orchestrating a spectrum of immune-associated diseases.
Collapse
Affiliation(s)
- Lijuan Fan
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Yaoyao Xia
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Youxia Wang
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Dandan Han
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Yanli Liu
- College of Animal Science and Technology, Northwest A&F University, Xi'an, 712100, China
| | - Jiahuan Li
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Jie Fu
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Leli Wang
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China
| | - Zhending Gan
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Bingnan Liu
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Jian Fu
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Congrui Zhu
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Zhenhua Wu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Jinbiao Zhao
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Hui Han
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - Hao Wu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Yiwen He
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, College of Life Sciences, Hunan Normal University, Changsha, 410081, China
| | - Yulong Tang
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China
| | - Qingzhuo Zhang
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Yibin Wang
- College of Animal Science and Technology, Northwest A&F University, Xi'an, 712100, China
| | - Fan Zhang
- College of Animal Science and Technology, Northwest A&F University, Xi'an, 712100, China
| | - Xin Zong
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China.
| | - Jie Yin
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, 410128, China.
| | - Xihong Zhou
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China.
| | - Xiaojun Yang
- College of Animal Science and Technology, Northwest A&F University, Xi'an, 712100, China.
| | - Junjun Wang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China.
| | - Yulong Yin
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China.
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, 410128, China.
| | - Wenkai Ren
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China.
| |
Collapse
|
4
|
Chen YK, Liu TT, Teia FKF, Xie MZ. Exploring the underlying mechanisms of obesity and diabetes and the potential of Traditional Chinese Medicine: an overview of the literature. Front Endocrinol (Lausanne) 2023; 14:1218880. [PMID: 37600709 PMCID: PMC10433171 DOI: 10.3389/fendo.2023.1218880] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 07/18/2023] [Indexed: 08/22/2023] Open
Abstract
Obesity and diabetes are closely related metabolic disorders that have become major public health concerns worldwide. Over the past few decades, numerous studies have explored the underlying mechanisms of these disorders and identified various risk factors, including genetics, lifestyle, and dietary habits. Traditional Chinese Medicine (TCM) has been increasingly recognized for its potential to manage obesity and diabetes. Weight loss is difficult to sustain, and several diabetic therapies, such as sulfonylureas, thiazolidinediones, and insulin, might make it harder to lose weight. While lifestyle changes should be the primary approach for people interested in lowering weight, drugs are also worth investigating. Since some of the newer glucose-lowering medications that cause weight loss, such as glucagon-like peptide-1 receptor agonists (GLP-1 RAs) and sodium-glucose cotransporter 2 inhibitors (SGLT2i), are additionally utilized or are under consideration for use as anti-obesity drugs, the frontier between glucose-lowering medication and weight loss drugs appears to be shifting. This review provides an overview of the literature on the underlying mechanisms of obesity and diabetes and the prospect of TCM in their management. We discuss the various TCM interventions, including acupuncture, herbal medicine, and dietary therapy, and their effects on metabolic health. We also highlight the potential of TCM in regulating gut microbiota, reducing inflammation, and improving insulin sensitivity. The findings suggest that TCM may provide a promising approach to preventing and managing obesity and diabetes. However, further well-designed studies are needed to confirm the efficacy and safety of TCM interventions and to elucidate their underlying mechanisms of action.
Collapse
Affiliation(s)
- Yan-kun Chen
- School of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
- Hunan Engineering Technology Research Center for Medicinal and Functional Food, Hunan University of Chinese Medicine, Changsha, China
- Key Laboratory of TCM Heart and Lung Syndrome Differentiation and Medicated Diet and Dietotherapy, Hunan University of Chinese Medicine, Changsha, China
| | - Ting-ting Liu
- School of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
- Hunan Engineering Technology Research Center for Medicinal and Functional Food, Hunan University of Chinese Medicine, Changsha, China
- Key Laboratory of TCM Heart and Lung Syndrome Differentiation and Medicated Diet and Dietotherapy, Hunan University of Chinese Medicine, Changsha, China
| | - Farah Khameis Farag Teia
- Department of Agro-technology, Medicinal and Aromatic Plants and Traditional Medicine Research Institute, National Centre for Research, Khartoum, Sudan
| | - Meng-zhou Xie
- School of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
- Hunan Engineering Technology Research Center for Medicinal and Functional Food, Hunan University of Chinese Medicine, Changsha, China
- Key Laboratory of TCM Heart and Lung Syndrome Differentiation and Medicated Diet and Dietotherapy, Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
5
|
Zhao Q, Wang Z, Wang X, Yan X, Guo Q, Yue Y, Yue T, Yuan Y. The bioaccessibility, bioavailability, bioactivity, and prebiotic effects of phenolic compounds from raw and solid-fermented mulberry leaves during in vitro digestion and colonic fermentation. Food Res Int 2023; 165:112493. [PMID: 36869449 DOI: 10.1016/j.foodres.2023.112493] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 12/19/2022] [Accepted: 01/20/2023] [Indexed: 01/26/2023]
Abstract
The bioaccessibility and bioactivity of phenolic compounds in mulberry leaves (MLs) relate to the digestion process. This study was aimed at investigating the release of phenolic compounds, as well as the potential bioactivities of raw MLs (UF-MLs) and solid-fermented MLs (F-MLs) during in vitro digestion and colonic fermentation. Antioxidant activities and phenolic compounds released in the digested extracts are shown in decreasing order of location: intestinal > oral > gastric. The bioavailability of total phenolics and flavonoids in F-MLs were 10.14 ± 1.81 % and 6.66 ± 0.55 %, respectively. There was no significant difference in the inhibitory activity of α-glucosidase during gastrointestinal digestion. For colonic fermentation, the highest free radical-scavenging ability of DPPH and ABTS was found at 24 h and 48 h, respectively. The release of phenolic compounds was not significantly different after 48 h of colonic fermentation. LC-MS/MS showed that liquiritigenin, apigenin, chlorogenic acid, and ferulic acid were the major compounds released in the small intestine digestion, and valerenic acid was the primary colonic metabolite. 16S rDNA showed that UF-MLs promoted the growth of Bifidobacterium and F-MLs lowered the Firmicutes-to-Bacteroidetes ratio. Furthermore, F-MLs increased the concentration of acetic acids (25.75 ± 0.86 mM) after 24 h of colonic fermentation. The results of this study indicated that F-MLs exhibit relatively higher phenolic bioaccessibility, antioxidant activities, and SCFA production and are a promising candidate as a health food supplement.
Collapse
Affiliation(s)
- Qiannan Zhao
- College of Food Science and Engineering, Northwest A & F University, Yangling 712100, China; Laboratory of Quality & Safety Risk Assessment for Agro-products (Yangling), Ministry of Agriculture, Yangling 712100, China
| | - Zewei Wang
- College of Food Science and Engineering, Northwest A & F University, Yangling 712100, China; Laboratory of Quality & Safety Risk Assessment for Agro-products (Yangling), Ministry of Agriculture, Yangling 712100, China
| | - Xin Wang
- College of Food Science and Engineering, Northwest A & F University, Yangling 712100, China; Laboratory of Quality & Safety Risk Assessment for Agro-products (Yangling), Ministry of Agriculture, Yangling 712100, China
| | - Xiaohai Yan
- College of Food Science and Engineering, Northwest A & F University, Yangling 712100, China; Laboratory of Quality & Safety Risk Assessment for Agro-products (Yangling), Ministry of Agriculture, Yangling 712100, China
| | - Qi Guo
- College of Food Science and Engineering, Northwest A & F University, Yangling 712100, China; Laboratory of Quality & Safety Risk Assessment for Agro-products (Yangling), Ministry of Agriculture, Yangling 712100, China
| | - Yuan Yue
- Xi'an Gaoxin, No. 1, High School, Xi'an 710,000, China
| | - Tianli Yue
- College of Food Science and Engineering, Northwest A & F University, Yangling 712100, China; Laboratory of Quality & Safety Risk Assessment for Agro-products (Yangling), Ministry of Agriculture, Yangling 712100, China; College of Food Science and Techonology, Northwest University, Xi'an 710069, China.
| | - Yahong Yuan
- College of Food Science and Engineering, Northwest A & F University, Yangling 712100, China; Laboratory of Quality & Safety Risk Assessment for Agro-products (Yangling), Ministry of Agriculture, Yangling 712100, China.
| |
Collapse
|
6
|
Liu Y, Yang J, Liu X, Liu R, Wang Y, Huang X, Li Y, Liu R, Yang X. Dietary folic acid addition reduces abdominal fat deposition mediated by alterations in gut microbiota and SCFA production in broilers. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2023; 12:54-62. [PMID: 36439290 PMCID: PMC9684696 DOI: 10.1016/j.aninu.2022.08.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 05/15/2022] [Accepted: 08/09/2022] [Indexed: 06/09/2023]
Abstract
Intensive selective breeding for high growth rate and body weight cause excess abdominal fat in broilers. Gut microbiota and folic acid were reported to regulate lipid metabolism. A total of 210 one-day-old broilers were divided into the control (folic acid at 1.3 mg/kg) and folic acid groups (folic acid at 13 mg/kg) to illustrate the effects of folic acid on growth performance, abdominal fat deposition, and gut microbiota, and the experiment lasted 28 d. Results revealed that dietary folic acid addition decreased abdominal fat percentage (P < 0.05) and down-regulated genes expression related to cell proliferation and differentiation in abdominal fat including IGF1, EGF, C/EBPα, PPARγ, PLIN1, FABP4 and PCNA (P < 0.05). Folic acid addition decreased caecal Firmicutes-to-Bacteroidetes ratio (P < 0.01) and increased the proportions of Alistipes, Oscillospira, Ruminococcus, Clostridium, Dehalobacterium and Parabacteroides (P < 0.05). Caecal acetic acid, and propionic acid contents were found to be higher under folic acid treatment (P < 0.05), which were negatively related to genes expression associated with adipocyte proliferation and differentiation (P < 0.05). Ruminococcus was positively correlated with caecal acetic acid content, and the same phenomenon was detected between propionic acid and Oscillospira and Ruminococcus (P < 0.05). Acetic acid and Oscillospira were identified to be negatively associated with abdominal fat percentage (P < 0.05). In conclusion, our data demonstrated that dietary supplementation of folic acid reduced fat deposition in broilers by inhibiting abdominal adipocyte proliferation and differentiation, which might be mediated by changes in gut microbiota and short chain fatty acid production.
Collapse
Affiliation(s)
- Yanli Liu
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Jiantao Yang
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Xiaoying Liu
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Rui Liu
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Yibin Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Xinhuo Huang
- Nano Vitamin Engineering Research Center of Shaanxi Province, Xi'an 710000, China
| | - Yingge Li
- Shaanxi Province Animal Husbandry Technology Extension Station, Xi'an 710016, China
| | - Ruifang Liu
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Xiaojun Yang
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| |
Collapse
|
7
|
Liu M, Shi W, Huang Y, Wu Y, Wu K. Intestinal flora: A new target for traditional Chinese medicine to improve lipid metabolism disorders. Front Pharmacol 2023; 14:1134430. [PMID: 36937840 PMCID: PMC10014879 DOI: 10.3389/fphar.2023.1134430] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 02/13/2023] [Indexed: 03/05/2023] Open
Abstract
Lipid metabolism disorders (LMD) can cause a series of metabolic diseases, including hyperlipidemia, obesity, non-alcoholic fatty liver disease (NAFLD) and atherosclerosis (AS). Its development is caused by more pathogenic factors, among which intestinal flora dysbiosis is considered to be an important pathogenic mechanism of LMD. In recent years, the research on intestinal flora has made great progress, opening up new perspectives on the occurrence and therapeutic effects of diseases. With its complex composition and wide range of targets, traditional Chinese medicine (TCM) is widely used to prevent and treat LMD. This review takes intestinal flora as a target, elaborates on the scientific connotation of TCM in the treatment of LMD, updates the therapeutic thinking of LMD, and provides a reference for clinical diagnosis and treatment.
Collapse
Affiliation(s)
- Min Liu
- Department of Gynecology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Wei Shi
- Department of Gynecology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Yefang Huang
- Department of Gynecology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Yeke Wu
- Department of Stomatology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Keming Wu
- Department of Gynecology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| |
Collapse
|
8
|
Wang W, Sun M, Yu J, Ma X, Han C. Relationship between Components, Intestinal Microbiota, and Mechanism of Hypoglycemic Effect of the Saggy Ink Cap Medicinal Mushroom (Coprinus Comatus, Agaricomycetes): A Review. Int J Med Mushrooms 2023; 25:81-90. [PMID: 37947066 DOI: 10.1615/intjmedmushrooms.2023050474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2023]
Abstract
Coprinus comatus is rich in a variety of nutrients, which has been reported to display a good hypoglycemic effect. However, there is no consensus on the hypoglycemic mechanism of this mushroom. Intestinal microbiota, a complex and intrinsic system, is closely related to metabolism. In this review, we discussed the potential relationship between certain components of C. comatus and intestinal microbiota to illustrate the possible hypoglycemic mechanism of C. comatus through intestinal microbiota. It will provide a new perspective for the study of hypoglycemic mechanism of C. comatus and promote the development and utilization of this mushroom.
Collapse
Affiliation(s)
- Wei Wang
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, P.R. China
| | - Min Sun
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, People's Republic of China
| | - Jinyan Yu
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, P.R. China
| | - Xumin Ma
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, P.R. China
| | - Chunchao Han
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, People's Republic of China; Shandong Provincial Collaborative Innovation Center for Quality Control and Construction of the Whole Industrial Chain of Traditional Chinese Medicine, Jinan, Shandong, 250355, People's Republic of China
| |
Collapse
|
9
|
Li D, Tang W, Wang Y, Gao Q, Zhang H, Zhang Y, Wang Y, Yang Y, Zhou Y, Zhang Y, Li H, Li S, Zhao H. An overview of traditional Chinese medicine affecting gut microbiota in obesity. Front Endocrinol (Lausanne) 2023; 14:1149751. [PMID: 36936157 PMCID: PMC10016694 DOI: 10.3389/fendo.2023.1149751] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 02/14/2023] [Indexed: 03/05/2023] Open
Abstract
Obesity, a chronic metabolic disease with a complex pathophysiology, is caused by several variables. High-fat diets lead to the disruption of the gut microbiota and impaired gut barrier function in obese people. The dysbiosis and its metabolites through the intestinal barrier lead to an imbalance in energy metabolism and inflammatory response, which eventually contributes to the development of chronic diseases such as diabetes, hypertension, and cardiovascular disease. Current medicines are therapeutic to obesity in the short term; however, they may bring significant physical and emotional problems to patients as major side effects. Therefore, it is urgent to explore new therapeutic methods that have definite efficacy, can be taken for a long time, and have mild adverse effects. Numerous studies have demonstrated that traditional Chinese medicine (TCM) can control the gut microbiota in a multi-targeted and comprehensive manner, thereby restoring flora homeostasis, repairing damaged intestinal mucosal barriers, and eventually curbing the development of obesity. The active ingredients and compounds of TCM can restore the normal physiological function of the intestinal mucosal barrier by regulating gut microbiota to regulate energy metabolism, inhibit fat accumulation, affect food appetite, and reduce intestinal mucosal inflammatory response, thereby effectively promoting weight loss and providing new strategies for obesity prevention and treatment. Although there are some studies on the regulation of gut microbiota by TCM to prevent and treat obesity, all of them have the disadvantage of being systematic and comprehensive. Therefore, this work comprehensively describes the molecular mechanism of obesity mediated by gut microbiota based on the research state of obesity, gut microbiota, and TCM. A comprehensive and systematic summary of TCM targeting the regulation of gut microbiota for the treatment of obesity should be conducted in order to provide new strategies and ideas for the treatment of obesity.
Collapse
Affiliation(s)
- Donghui Li
- College of Pharmacy, Jiamusi University, Jiamusi, China
| | - Weiwei Tang
- College of Pharmacy, Jiamusi University, Jiamusi, China
| | - Yanyan Wang
- College of Pharmacy, Jiamusi University, Jiamusi, China
| | - Qi Gao
- College of Pharmacy, Jiamusi University, Jiamusi, China
| | - Hongwei Zhang
- Department of Emergency Surgery, The First Affiliated Hospital of Jiamusi University, Jiamusi, China
| | - Yu Zhang
- College of Pharmacy, Jiamusi University, Jiamusi, China
| | - Yuliang Wang
- College of Pharmacy, Jiamusi University, Jiamusi, China
| | - Yongyi Yang
- College of Pharmacy, Jiamusi University, Jiamusi, China
| | - Yingming Zhou
- College of Pharmacy, Jiamusi University, Jiamusi, China
| | - Yike Zhang
- College of Pharmacy, Jiamusi University, Jiamusi, China
| | - Haonan Li
- College of Pharmacy, Jiamusi University, Jiamusi, China
| | - Shuo Li
- College of Pharmacy, Jiamusi University, Jiamusi, China
| | - Hong Zhao
- College of Pharmacy, Jiamusi University, Jiamusi, China
- *Correspondence: Hong Zhao,
| |
Collapse
|
10
|
Lee DPS, Peng A, Taniasuri F, Tan D, Kim JE. Impact of fiber-fortified food consumption on anthropometric measurements and cardiometabolic outcomes: A systematic review, meta-analyses, and meta-regressions of randomized controlled trials. Crit Rev Food Sci Nutr 2022; 63:8301-8319. [PMID: 35333681 DOI: 10.1080/10408398.2022.2053658] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The consumption of processed and refined food lacking in fiber has led to global prevalence of obesity and cardiometabolic diseases. Fiber-fortification into these foods can yield potential health improvements to reduce disease risk. This meta-analyses aimed to evaluate how fiber-fortified food consumption changes body composition, blood pressure, blood lipid-lipoprotein panel, and glycemic-related markers. Searches were performed from 5 databases, with 31 randomized controlled trial eventually analyzed. Hedges' g values (95% confidence interval [CI]) attained from outcome change values were calculated using random-effects model. Fiber-fortified food significantly reduced body weight (-0.31 [-0.59, -0.03]), fat mass (-0.49 [-0.72, -0.26]), total cholesterol (-0.54 [-0.71, -0.36]), low-density lipoprotein cholesterol (-0.49 [-0.65, -0.33]), triglycerides (-0.24 [-0.36, -0.12]), fasting glucose (-0.30 [-0.49, -0.12]), and HbA1c (-0.44 [-0.74, -0.13]). Subgroup analysis differentiated soluble fiber as significantly reducing triglycerides and insulin while insoluble fiber significantly reduced body weight, BMI, and HbA1c. Greater outcome improvements were observed with solid/semi-solid food state than liquid state. Additionally, fiber fortification of <15 g/day induced more health outcome benefits compared to ≥15 g/day, although meta-regression found a dose-dependent improvement to waist circumference (p-value = 0.036). Findings from this study suggest that consuming food fortified with dietary fiber can improve anthropometric and cardiometabolic outcomes.Supplemental data for this article is available online at https://doi.org/10.1080/10408398.2022.2053658.
Collapse
Affiliation(s)
- Delia Pei Shan Lee
- Department of Food Science and Technology, National University of Singapore, Singapore, Singapore
| | - Aiwei Peng
- Department of Food Science and Technology, National University of Singapore, Singapore, Singapore
| | - Fransisca Taniasuri
- Department of Food Science and Technology, National University of Singapore, Singapore, Singapore
| | - Denise Tan
- Department of Food Science and Technology, National University of Singapore, Singapore, Singapore
- Science and Technology Department, Nestlé R&D Center (Pte) Ltd, Singapore, Singapore
| | - Jung Eun Kim
- Department of Food Science and Technology, National University of Singapore, Singapore, Singapore
| |
Collapse
|
11
|
Shi H, Ter Horst R, Nielen S, Bloemendaal M, Jaeger M, Joosten I, Koenen H, Joosten LAB, Schweren LJS, Vasquez AA, Netea MG, Buitelaar J. The gut microbiome as mediator between diet and its impact on immune function. Sci Rep 2022; 12:5149. [PMID: 35338162 PMCID: PMC8956630 DOI: 10.1038/s41598-022-08544-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 02/18/2022] [Indexed: 12/22/2022] Open
Abstract
Dietary habits may affect inflammatory status in humans. Here we explore this interaction as well as the potential mediating role of the gut microbiome (GM), given that the GM is both involved in processing of dietary components and influences the immune system. A cross-sectional analysis of a sample of 482 healthy participants (207 males and 275 females) was performed. Dietary intake was assessed by a semiquantitative food questionnaire. Adipokines and soluble inflammatory mediators were assayed with multiple immunoassays and ELISA. Microbial DNA was extracted from frozen stool samples of 471 participants. Polychoric correlation analysis was used to establish dietary patterns, and joint multivariate associations between these dietary patterns and immune biomarkers were studied using regression analyses with adjustment for sex, age, BMI, smoking, education levels and physical exercise and other dietary patterns. Non-parametric entropy mediation was applied to investigate whether diet-immune relationships are mediated by abundance of microbial species. In this cohort, we identified three dietary patterns, characterized as “high-meat” (meat and sweetened drink), “prudent diet” (fish, fruit, legumes and vegetables) and “high alcohol” (higher alcohol consumption). Higher adherence to prudent diet was associated with a higher adiponectin level. The high alcohol pattern was associated with high concentrations of circulating concentrations of pro-inflammatory markers (CRP, IL-6, VEGF). Dialister invisus was found to mediate the relationship between a prudent dietary pattern and adiponectin, AAT, CRP, IL-6, and VEGF. In conclusion, a meat-based diet and a diet with high alcohol consumption were associated with high concentrations of biomarkers of chronic low-grade inflammation, and conversely, a prudent diet was associated with anti-inflammatory biomarkers. Diet-inflammation regulation may differ between sexes. Mediation analyses revealed that the association between prudent diet and immune function was partially mediated by the GM. The study adds to our understanding of the associations between diet, the immune system and the GM in a healthy population.
Collapse
Affiliation(s)
- Huiqing Shi
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Radboud University Nijmegen Medical Centre, Radboudumc, Kapittelweg 29, 6525 EN, Nijmegen, Gelderland, The Netherlands.
| | - Rob Ter Horst
- Department of Internal Medicine, Radboudumc Center for Infectious Diseases, Radboudumc, 6500 HB, Nijmegen, Gelderland, The Netherlands
| | - Suzanne Nielen
- Department of Psychiatry, and Department of Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboudumc, 6525 EN, Nijmegen, Gelderland, The Netherlands
| | - Mirjam Bloemendaal
- Department of Psychiatry, and Department of Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboudumc, 6525 EN, Nijmegen, Gelderland, The Netherlands
| | - Martin Jaeger
- Department of Internal Medicine, Radboudumc Center for Infectious Diseases, Radboudumc, 6500 HB, Nijmegen, Gelderland, The Netherlands
| | - Irma Joosten
- Department of Laboratory Medicine, Radboudumc, 6500 HB, Nijmegen, Gelderland, The Netherlands
| | - Hans Koenen
- Department of Laboratory Medicine, Radboudumc, 6500 HB, Nijmegen, Gelderland, The Netherlands
| | - Leo A B Joosten
- Department of Internal Medicine, Radboudumc Center for Infectious Diseases, Radboudumc, 6500 HB, Nijmegen, Gelderland, The Netherlands
| | - Lizanne J S Schweren
- Interdisciplinary Center Psychopathology and Emotion Regulation, University Medical Center Groningen, University of Groningen, 9700 RB, Hanzeplein 1, Groningen, The Netherlands
| | - Alejandro Arias Vasquez
- Department of Psychiatry, and Department of Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboudumc, 6525 EN, Nijmegen, Gelderland, The Netherlands
| | - Mihai G Netea
- Department of Internal Medicine, Radboudumc Center for Infectious Diseases, Radboudumc, 6500 HB, Nijmegen, Gelderland, The Netherlands.,Department of Immunology and Metabolism, Life and Medical Sciences Institute (LIMES), University of Bonn, Bonn, Germany
| | - Jan Buitelaar
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Radboud University Nijmegen Medical Centre, Radboudumc, Kapittelweg 29, 6525 EN, Nijmegen, Gelderland, The Netherlands.,Karakter Child and Adolescent Psychiatry University Centre, 6525 GC, Nijmegen, Gelderland, The Netherlands
| |
Collapse
|
12
|
Zhang L, Liu C, Jiang Q, Yin Y. Butyrate in Energy Metabolism: There Is Still More to Learn. Trends Endocrinol Metab 2021; 32:159-169. [PMID: 33461886 DOI: 10.1016/j.tem.2020.12.003] [Citation(s) in RCA: 152] [Impact Index Per Article: 50.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 12/18/2020] [Accepted: 12/20/2020] [Indexed: 12/25/2022]
Abstract
Butyrate, a main product of gut microbial fermentation, has been recognized as an important mediator of gut microbiota regulation in whole body energy homeostasis. However, the mechanisms of butyrate metabolic control remain unclear. This review summarizes studies that directly examined the effects of butyrate on metabolic health. The effects of butyrate on metabolic functions, including thermogenesis, lipid and glucose metabolism, appetite, inflammation, and influence on gut microbiota, are described. The effects of butyrate on cellular systems via G protein-coupled receptors (GPRs), as a histone deacetylase inhibitor, and as a substrate that is metabolized intercellularly, are also discussed. Hopefully, a better understanding of butyrate metabolic regulation may provide new perspectives for the nutritional prevention and treatment of metabolic diseases.
Collapse
Affiliation(s)
- Lin Zhang
- Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Chudan Liu
- Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Qingyan Jiang
- Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, 510642, China.
| | - Yulong Yin
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Laboratory of Animal Nutritional Physiology and Metabolic Process, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan 410125, China.
| |
Collapse
|
13
|
Guan B, Wang L, Ma L, Liu X, Liu L. EFFECTS OF EXENDIN-4 ON ENDOPLASMIC RETICULUM STRESS-MEDIATED INSULIN RESISTANCE IN 3T3-L1 ADIPOCYTES. ACTA ENDOCRINOLOGICA-BUCHAREST 2019; 15:158-164. [PMID: 31508171 DOI: 10.4183/aeb.2019.158] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Objective Endoplasmic reticulum stress (ERS) is suspected as an important factor in the initiation of insulin resistance. Aim To explore the effects of exendin-4 (Ex-4) on the endoplasmic reticulum stress (ERS)-mediated insulin resistance in 3T3-L1 adipocytes. In our study, 3T3-L1 adipocytes were pre-treated with ERS inhibitors tauroursodeoxycholic acid (TUDCA), Ex-4 and an ERS inducer tunicamycin (TM) then induced insulin resistance. Glucose consumption of the adipocytes was measured. Western blots determined the protein levels of ERS markers and insulin signaling pathway. Results TM treatment reduced insulin-stimulated glucose consumption by 19.7% in 3T3-L1 adipocytes. This repression was blunted by 24h pre-treatment with TUDCA or Ex-4. Ex-4 augmented insulin-stimulated glucose consumption in adipocytes by 14.9%. Western blotting showed that TM treatment significantly increased the ER stress markers including p-IRE, p-JNK, p-PERK, p-eIF2a and ATF6 expression, whereas 24h pre-treatment of adipocytes with TUDCA or Ex-4 alleviated the ER stress. Ex-4 alleviates ERS-induced insulin resistance by upregulating the expression of phosphorylated Akt. Conclusion ERs mediates insulin resistance in 3T3-L1 adipocytes, and exendin-4 significantly improves this insulin resistance.
Collapse
Affiliation(s)
- B Guan
- Dept. of Endocrinology, Union Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - L Wang
- Dept. of Endocrinology, Union Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - L Ma
- Dept. of Endocrinology, Union Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - X Liu
- Dept. of Endocrinology, Union Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - L Liu
- Dept. of Endocrinology, Union Hospital, Fujian Medical University, Fuzhou, Fujian, China
| |
Collapse
|
14
|
Berkoz M. EFFECT OF HYPEROSIDE ON THE INHIBITION OF ADIPOGENESIS IN 3T3-L1 ADIPOCYTES. ACTA ENDOCRINOLOGICA-BUCHAREST 2019; 15:165-172. [PMID: 31508172 DOI: 10.4183/aeb.2019.165] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Context The inhibition of adipocyte differentiation has a significant role on the prevention of obesity and obesity-associated complications. Objective In this study, we aimed to detect whether hyperoside is able to inhibit the conversion of pre-adiposits into mature adiposits. Design and Methods 3T3-L1 pre-adipocytes were stimulated so as to differentiate into mature adipocytes. Hyperoside in non-cytotoxic concentrations (1, 2, 5, and 10 µM) were separately applied to differentiated 3T3-L1 cells. Oil red O staining was performed and triacylglycerol contents were measured. Furthermore, gene and protein expressions of transcription factors, adipogenic genes and adipokines were examined in order to investigate the effect of hyperoside on adipocyte differentiation. Results Hyperoside in high concentrations significantly suppressed the adipogenic process by inhibiting the expression of transcription factors and adipogenic genes and reducing lipid accumulation in adipocytes (p<0.05). Low doses of hyperoside are able to inhibit adipogenesis, but higher doses are needed to reduce fat accumulation in mature adipocytes. In the case of maturing preadipocytes, 5 μM of hyperoside exerts its antiadipogenic effect at the early stages of adipogenesis, whereas 10 μM of hyperoside acts at the later stages (p<0.05). Conclusion These results suggest that hyperoside has a beneficial effect on the prevention and treatment of obesity.
Collapse
Affiliation(s)
- M Berkoz
- Yuzuncu Yil University, Faculty of Pharmacy - Department of Biochemistry, Van, Turkey
| |
Collapse
|