1
|
Zhang C, Chen S, Wang Z, Zhang J, Yu W, Wang Y, Si W, Zhang Y, Zhang Y, Liang T. Exploring the mechanism of intestinal bacterial translocation after severe acute pancreatitis: the role of Toll-like receptor 5. Gut Microbes 2025; 17:2489768. [PMID: 40243695 PMCID: PMC11980482 DOI: 10.1080/19490976.2025.2489768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Revised: 03/31/2025] [Accepted: 04/01/2025] [Indexed: 04/18/2025] Open
Abstract
Severe acute pancreatitis (SAP)-induced intestinal bacterial translocation and enterogenic infection are among the leading causes of mortality in patients. However, the mechanisms by which SAP disrupted the intestinal barrier and led to bacterial translocation remained unclear. Therefore, we employed multi-omics analysis including microbiome, metabolome, epigenome, transcriptome, and mass cytometry (CyTOF) to identify potential targets, followed by functional validation using transgenic mice. The integrated multi-omics analysis primarily indicated overgrowth of intestinal flagellated bacteria, upregulation of intestinal Toll-like receptor 5 (TLR5) and acute inflammatory response, and increased infiltration of intestinal high-expressing TLR5 lamina propria dendritic cells (TLR5hi LPDC) after SAP. Subsequently, intestinal flagellin-TLR5 signaling was activated after SAP. Intestinal barrier disruption, bacterial translocation, and helper T cells (Th) differentiation imbalance caused by SAP were alleviated in TLR5 knocked out (Tlr5-/-) or conditionally knocked out on LPDC (Tlr5ΔDC) mice. However, TLR5 conditional knockout on intestinal epithelial cells (Tlr5ΔIEC) failed to improve SAP-induced bacterial translocation. Moreover, depletion of LPDC and regulatory T cells (Treg) ameliorated bacterial translocation after SAP. Our findings identify TLR5 on LPDC as a potential novel target for preventing or treating intestinal bacterial translocation caused by SAP.
Collapse
Affiliation(s)
- Cheng Zhang
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, Zhejiang Province, China
- Innovation Center for the Study of Pancreatic Diseases, Hangzhou, Zhejiang Province, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, Zhejiang Province, China
| | - Shiyin Chen
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, Zhejiang Province, China
- Innovation Center for the Study of Pancreatic Diseases, Hangzhou, Zhejiang Province, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, Zhejiang Province, China
| | - Zhien Wang
- Department of Rehabilitation, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Jian Zhang
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, Zhejiang Province, China
- Innovation Center for the Study of Pancreatic Diseases, Hangzhou, Zhejiang Province, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, Zhejiang Province, China
| | - Wenqiao Yu
- Department of Surgical Intensive Care Unit, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Yanshuai Wang
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, Zhejiang Province, China
- Innovation Center for the Study of Pancreatic Diseases, Hangzhou, Zhejiang Province, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, Zhejiang Province, China
| | - Weiwei Si
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, Zhejiang Province, China
- Innovation Center for the Study of Pancreatic Diseases, Hangzhou, Zhejiang Province, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, Zhejiang Province, China
| | - Yuwei Zhang
- College of Science, Mathematics and Technology, Wenzhou-Kean University, Wenzhou, Zhejiang Province, China
| | - Yun Zhang
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, Zhejiang Province, China
- Innovation Center for the Study of Pancreatic Diseases, Hangzhou, Zhejiang Province, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, Zhejiang Province, China
- Department of Nutrition, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Tingbo Liang
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, Zhejiang Province, China
- Innovation Center for the Study of Pancreatic Diseases, Hangzhou, Zhejiang Province, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, Zhejiang Province, China
| |
Collapse
|
2
|
Li J, Bi D, Nie Y, Wu H, Lei Y, Yu S, Rong H, Yang Y, Lei Z. Loperamide-induced constipation is associated with excessive accumulation of bile acids and cholesterol in the liver of mice; attenuation by hesperidin. Food Chem Toxicol 2025:115561. [PMID: 40389132 DOI: 10.1016/j.fct.2025.115561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2025] [Revised: 05/16/2025] [Accepted: 05/16/2025] [Indexed: 05/21/2025]
Abstract
Loperamide, a widely used antidiarrheal agent, frequently induces constipation alongside other adverse effects. This study explored hesperidin's therapeutic potential in alleviating loperamide-induced constipation and its underlying mechanisms. Constipation models were established in HFD- or NFD-fed mice via loperamide administration (5 mg/kg/day). Hesperidin (100 mg/kg/day) significantly increased fecal weight and moisture in constipated mice. Biochemical analyses revealed elevated cholic acid/chenodeoxycholic acid ratios in serum and liver tissues of loperamide-treated HFD mice, indicating bile acids dysregulation. qRT-PCR and Western blot results demonstrated that hesperidin downregulated hepatic expression of cholesterol/bile acids biosynthesis genes (e.g., Hmgcr, Cyp7a1, Ch25h), which were overexpressed in constipated mice. Concurrently, hesperidin enhanced the expression of transporters (Abcg5, Abcb11, Abcc2) responsible for biliary cholesterol and bile acids efflux. Furthermore, hesperidin upregulated hepatic nuclear receptors (FXR/SHP), key regulators of bile acids homeostasis. Hesperidin alleviates loperamide-induced constipation through two parallel mechanisms: decreasing hepatic cholesterol accumulation and enhancing bile acids excretion. This dual action results from coordinated regulation of biosynthesis enzymes and transport proteins. This study highlights hesperidin's potential as an adjunct therapy to counteract loperamide-related constipation.
Collapse
Affiliation(s)
- Jiahui Li
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China; School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou Higher Education Mega Center, Guangzhou 510006, P.R. China
| | - Dan Bi
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Ya Nie
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China; School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou Higher Education Mega Center, Guangzhou 510006, P.R. China
| | - Huijuan Wu
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China; School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou Higher Education Mega Center, Guangzhou 510006, P.R. China
| | - Yuting Lei
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China
| | - Siping Yu
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China; School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou Higher Education Mega Center, Guangzhou 510006, P.R. China
| | - Hedong Rong
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China; School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou Higher Education Mega Center, Guangzhou 510006, P.R. China
| | - Yanhong Yang
- The First Affiliated Hospital (School of Clinical Medicine), Guangdong Pharmaceutical University, Nong-Lin-Xia Road 19#, Yue-Xiu District, Guangzhou 510080, P.R. China.
| | - Zili Lei
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China.
| |
Collapse
|
3
|
Li S, Tan H, Yang J, Yao H, Nie X, Peng X, Liu Q, Yang W, Liu G, Nie Q, Bian S, Huang X, Yin JY, Cui SW, Nie SP. Effects of Three Homogalacturonan-Type Pectins on Mice with Metabolic Syndrome. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025. [PMID: 40376805 DOI: 10.1021/acs.jafc.4c11988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2025]
Abstract
Pectin, a class of dietary fiber, has received increasing attention in recent years for its ameliorative effects on metabolic diseases. However, the structural variability of pectin leads to differential effects on these diseases. The intrinsic mechanism by which pectin, derived from different sources, differentially influences metabolic syndrome by interacting with gut microbiota and host metabolism remains elusive and warrants thorough investigation. To address this, we investigated the effects of HG-type pectins from apple, citrus, and pomelo on phenotypic expressions, inflammatory factors, oxidative stress, and serum hormone levels in mice with metabolic syndrome. In addition, we sought to identify pivotal bacterial species and metabolites by integrating genomics and metabolomics approaches. Our exploration also extended to the relationship between structural characteristics of pectins, gut microbiota, and metabolic syndrome. Our findings revealed that the three pectins diversely improved metabolic syndrome in mice, which correlated with gut microbiota and their beneficial metabolites. Notably, all three pectins were closely associated with Bacteroides and Bacteroides acidifaciens. Besides, the potential mediators of the therapeutic effects included Bacteroides, Lactococcus, and Lachnoclostriclum for apple pectin; Colidextribacter, Bacteroides, Lachnospiraceae_NK4A136_group, and Lachnoclostriclum for citrus pectin; and Lachnospiraceae_NK4A136_group, Bacteroides, and Mucispirillum for pomelo pectin. Metabolites such as arachidonic acid, kynurenic acid, lithocholic acid, deoxycholic acid, and indoleacetic acid, linked to these microbes, may serve as the mediators of pectin's benefits. Ultimately, the molecular weight, degree of esterification, and monosaccharide composition of pectins significantly influenced the outcomes. This study may contribute to a more nuanced understanding that can inform targeted nutritional strategies to modulate gut microbiota for metabolic syndrome management.
Collapse
Affiliation(s)
- Song Li
- State Key Laboratory of Food Science and Resources, China-Canada Joint Laboratory of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang 330047, China
| | - Huizi Tan
- State Key Laboratory of Food Science and Resources, China-Canada Joint Laboratory of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang 330047, China
| | - Jingrui Yang
- State Key Laboratory of Food Science and Resources, China-Canada Joint Laboratory of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang 330047, China
| | - Hong Yao
- State Key Laboratory of Food Science and Resources, China-Canada Joint Laboratory of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang 330047, China
| | - Xinke Nie
- State Key Laboratory of Food Science and Resources, China-Canada Joint Laboratory of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang 330047, China
| | - Xiaomao Peng
- State Key Laboratory of Food Science and Resources, China-Canada Joint Laboratory of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang 330047, China
| | - Qionglian Liu
- State Key Laboratory of Food Science and Resources, China-Canada Joint Laboratory of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang 330047, China
| | - Wanyu Yang
- State Key Laboratory of Food Science and Resources, China-Canada Joint Laboratory of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang 330047, China
| | - Guohui Liu
- State Key Laboratory of Food Science and Resources, China-Canada Joint Laboratory of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang 330047, China
| | - Qixing Nie
- State Key Laboratory of Food Science and Resources, China-Canada Joint Laboratory of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang 330047, China
| | - Shuigen Bian
- State Key Laboratory of Food Science and Resources, China-Canada Joint Laboratory of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang 330047, China
| | - Xiaojun Huang
- State Key Laboratory of Food Science and Resources, China-Canada Joint Laboratory of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang 330047, China
| | - Jun-Yi Yin
- State Key Laboratory of Food Science and Resources, China-Canada Joint Laboratory of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang 330047, China
| | - Steve W Cui
- State Key Laboratory of Food Science and Resources, China-Canada Joint Laboratory of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang 330047, China
- Guelph Research and Development Centre, Agriculture and Agri-Food Canada, 93 Stone Road West, Guelph, ON N1G 5C9, Canada
| | - Shao-Ping Nie
- State Key Laboratory of Food Science and Resources, China-Canada Joint Laboratory of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang 330047, China
| |
Collapse
|
4
|
Su FZ, Bai CX, Zhang WS, Liu M, Li B, Sun MH, He YJ, Zeng YN, Sun YP, Yang BY, Kuang HX, Wang QH. Lipid-lowering effects of bile Arisaema polysaccharides on high-fat diet-induced hyperlipidemia: An integrated analysis of metabolomics, lipidomics and microbiome. Int J Biol Macromol 2025; 311:143932. [PMID: 40348213 DOI: 10.1016/j.ijbiomac.2025.143932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 04/22/2025] [Accepted: 05/03/2025] [Indexed: 05/14/2025]
Abstract
Bile Arisaema, a traditional Chinese medicine, has been previously identified by our team to possess antipyretic properties attributed to its polysaccharide component. Recently, we have confirmed that bile Arisaema played a significantly lipid-lowering effect on hyperlipidemia rats. Building upon this discovery, the present study aimed to investigate the unexplored hypolipidemic potential of its polysaccharide component and elucidate the underlying mechanisms. A soluble polysaccharide fraction devoid of free proteins, named BAPs, was extracted from bile Arisaema using a combination of hot water extraction, alcohol precipitation, and the Sevage method. The structural characteristics of BAPs were preliminarily elucidated through monosaccharide composition analysis (mainly composed of glucose), molecular weight distribution (38.74 kDa and 2.87 kDa), and glycosyl linkage analysis via methylation. The results of animal experiment demonstrated that oral administration of BAPs (400 mg/kg/day) for four weeks significantly improved abnormal serum lipid levels, hepatic function and histopathological injury on high-fat diet-induced hyperlipidemia rats. Mechanistically, the results of high throughput sequencing indicated that BAPs intake markedly altered the hepatic and fecal metabolome and lipidome, while also modulating gut microbiota composition and improving intestinal barrier integrity. Spearman's correlation analysis unveiled closely associations between the altered microbes, lipids, metabolites and serum biochemical indicators. Western blotting and qRT-PCR analyses further confirmed that these metabolic improvements were mediated by the regulation of key genes involved in lipid metabolism. Collectively, this study demonstrated that BAP supplementation effectively improved serum lipid profiles in hyperlipidemia rats by modulating metabolic disorders and restoring gut homeostasis.
Collapse
Affiliation(s)
- Fa-Zhi Su
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin 150040, China
| | - Chen-Xi Bai
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin 150040, China
| | - Wen-Sen Zhang
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin 150040, China
| | - Meng Liu
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin 150040, China
| | - Biao Li
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin 150040, China
| | - Ming-Hao Sun
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin 150040, China
| | - Yu-Jia He
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin 150040, China
| | - Yuan-Ning Zeng
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Yan-Ping Sun
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin 150040, China
| | - Bing-You Yang
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin 150040, China
| | - Hai-Xue Kuang
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin 150040, China.
| | - Qiu-Hong Wang
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin 150040, China; School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| |
Collapse
|
5
|
Zhang J, Zhou J, He Z, Xia Z, Liu H, Wu Y, Chen S, Wu B, Li H. Salidroside attenuates NASH through regulating bile acid-FXR/TGR5 signaling pathway via targeting gut microbiota. Int J Biol Macromol 2025; 307:142276. [PMID: 40118401 DOI: 10.1016/j.ijbiomac.2025.142276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 02/15/2025] [Accepted: 03/17/2025] [Indexed: 03/23/2025]
Abstract
Nonalcoholic steatohepatitis (NASH) is a significant threat to human health. Our previous study revealed that salidroside attenuated NASH and regulated the gut microbiota. However, whether the therapeutic effect of salidroside depends on gut microbiota remains to be determined. Therefore, we conducted further experiments to elucidate the essential functions of gut microbiota-associated metabolic pathways in the anti-NASH effects of salidroside. Our results showed that salidroside effectively alleviated lipid accumulation and inflammatory injury in NASH mice. 16S rRNA sequencing revealed that salidroside increased the abundance of Bacteroides. Mice receiving fecal microbiota transplantation (FMT) from salidroside-treated also presented less hepatic steatosis and higher abundance of Bacteroides. Antibiotics eliminated the effects of salidroside on hepatic steatosis and the gut microbiota. Mechanistically, salidroside and FMT from salidroside-treated altered the bile acid (BA) profile by decreasing the levels of conjugated BAs and tauro-α/β-muricholic acid and activated downstream farnesoid X receptor (FXR) and Takeda G protein-coupled receptor 5 (TGR5). Furthermore, we found that inhibitors of bile salt hydrolase (BSH) and FXR/TGR5 abolished the effects of salidroside and reduced downstream carnitine palmitoyltransferase 1α and lipoprotein lipase expression. These data demonstrate that salidroside attenuated NASH via gut microbiota-BA-FXR/TGR5 signaling pathway and reveal the underlying mechanism of salidroside on NASH.
Collapse
Affiliation(s)
- Jun Zhang
- Liver Disease Department of Integrative Medicine, Ningbo No.2 Hospital, Ningbo, Zhejiang 315000, China; Cixi Biomedical Research Institute, Wenzhou Medical University, Ningbo, Zhejiang 315300, China
| | - Jing Zhou
- Liver Disease Department of Integrative Medicine, Ningbo No.2 Hospital, Ningbo, Zhejiang 315000, China
| | - Zheyun He
- Liver Diseases Institute, Ningbo No. 2 Hospital, Ningbo, Zhejiang 315000, China
| | - Zhanyang Xia
- Liver Disease Department of Integrative Medicine, Ningbo No.2 Hospital, Ningbo, Zhejiang 315000, China
| | - Hongliang Liu
- Liver Disease Department of Integrative Medicine, Ningbo No.2 Hospital, Ningbo, Zhejiang 315000, China
| | - Yuan Wu
- Liver Disease Department of Integrative Medicine, Ningbo No.2 Hospital, Ningbo, Zhejiang 315000, China
| | - Si Chen
- Liver Disease Department of Integrative Medicine, Ningbo No.2 Hospital, Ningbo, Zhejiang 315000, China
| | - Boming Wu
- Liver Disease Department of Integrative Medicine, Ningbo No.2 Hospital, Ningbo, Zhejiang 315000, China
| | - Hongshan Li
- Liver Disease Department of Integrative Medicine, Ningbo No.2 Hospital, Ningbo, Zhejiang 315000, China; Medical Experimental Department of Ningbo No.2 Hospital, Ningbo, Zhejiang 315000, China.
| |
Collapse
|
6
|
Pan H, Su J, Bai M, Chen M, He Z, Rong N, Lin X, Wang H, Wu H. Two Caffeoyl-CoA O-methyltransferase-like enzyme are involved in the biosynthesis of polymethoxyflavones in Citrus reticulata 'Chachiensis'. Int J Biol Macromol 2025; 310:143277. [PMID: 40288725 DOI: 10.1016/j.ijbiomac.2025.143277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 04/10/2025] [Accepted: 04/16/2025] [Indexed: 04/29/2025]
Abstract
Polymethoxyflavones (PMFs) are abundant in citrus plants and exhibit remarkable biological activities in the inhibition of the occurrence and development of various tumours. O-methyltransferase (OMTs)-mediated methylation plays a crucial role in the biosynthesis of PMFs. However, the OMTs catalyzing the methylation of hydroxyflavonoids in citrus have not been fully identified. In this study, we identified two novel CCoAOMT-like enzymes in Citrus reticulata 'Chachiensis', designated CrcCCoAOMT7-1 and CrcCCoAOMT7-2. The transient overexpression and virus-induced gene silencing experiments further confirmed the essential role of CrcCCoAOMT7-1/7-2 in the regulation of PMF synthesis in citrus. Furthermore, in vitro functional analysis showed that CrcCCoAOMT7-1 preferentially catalyzes flavones, while CrcCCoAOMT7-2 exhibits a preference for flavanones and quercetin (flavonol). The two enzymes methylate the 3'-, 4'-, 3-, 6-, 8-OH sites and the 3'-, 4'-, 6-, 7-, 8-OH sites with hydroxyflavonoids, respectively, covering nearly all methylation sites during the biosynthesis of PMFs. Functional diversification of CCoAOMTs expended our understanding of PMFs biosynthesis modes in citrus. This study enriches the PMF biosynthetic pathway in citrus and lays the foundation for the selection and breeding of high-quality Citrus reticulata 'Chachiensis' mandarins, as well as the synthesis of natural anticancer PMFs through genetic engineering.
Collapse
Affiliation(s)
- Huimin Pan
- Centre for Medicinal Plant Research, College of Life Sciences, South China Agricultural University, Guangzhou 510642, China
| | - Jianmu Su
- Centre for Medicinal Plant Research, College of Life Sciences, South China Agricultural University, Guangzhou 510642, China
| | - Mei Bai
- Centre for Medicinal Plant Research, College of Life Sciences, South China Agricultural University, Guangzhou 510642, China
| | - Meizhuang Chen
- Centre for Medicinal Plant Research, College of Life Sciences, South China Agricultural University, Guangzhou 510642, China
| | - Zhuoyuan He
- Centre for Medicinal Plant Research, College of Life Sciences, South China Agricultural University, Guangzhou 510642, China
| | - Ning Rong
- Centre for Medicinal Plant Research, College of Life Sciences, South China Agricultural University, Guangzhou 510642, China
| | - Xitong Lin
- Centre for Medicinal Plant Research, College of Life Sciences, South China Agricultural University, Guangzhou 510642, China
| | - Haiyang Wang
- Yazhouwan National Laboratory, Sanya 572024, China
| | - Hong Wu
- Centre for Medicinal Plant Research, College of Life Sciences, South China Agricultural University, Guangzhou 510642, China.
| |
Collapse
|
7
|
Ding C, Ruan J, Huang J, Liu L, Li Y, Du Y, Zhao Y. Nuciferine activates intestinal TAS2R46 to attenuate metabolic disorders and hyperlipidemia via hepatic VLDL regulation. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 142:156800. [PMID: 40311594 DOI: 10.1016/j.phymed.2025.156800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 04/10/2025] [Accepted: 04/21/2025] [Indexed: 05/03/2025]
Abstract
BACKGROUND Dysregulated blood lipid metabolism, a primary driver of hyperlipidemia, is closely associated with excessive very low-density lipoprotein (VLDL) synthesis and secretion. Nuciferine, a bioactive compound isolated from lotus leaves, demonstrates remarkable hypolipidemic efficacy; however, its limited bioavailability challenges existing mechanistic explanations for this pronounced therapeutic effect. PURPOSE This research aims to investigate the disease-modifying effect and underlying mechanism of nuciferine against hyperlipidemia from a novel perspective by modulating the synthesis and secretion of VLDL. METHODS High-fat diet-induced hyperlipidemic rats were assessed by biochemical assays and histopathological examinations to assess the therapeutic effect of nuciferine. Untargeted metabo-lipidomics were launched to obtain the metabolic and lipid profiles, and explainable machine learning algorithms were innovatively utilized in screening differentially expressed metabolites for pathway analysis. A hyperlipidemic two-layer cell co-culture model was analyzed using quantitative polymerase chain reaction, molecular docking, immunofluorescence, cellular thermal shift assay, western blotting, and flow cytometry to delineate VLDL regulatory mechanisms. RESULTS Nuciferine significantly attenuated lipid accumulation and metabolic dysfunction in a hyperlipidemic rat model, with the biosynthesis and metabolism of phenylalanine, tyrosine, and tryptophan as pivotal metabolic pathways. Mechanistically, nuciferine activated intestinal type 2 taste receptor 46 (TAS2R46), promoting Ca²⁺-dependent secretion of glucagon-like peptide-1 (GLP-1). Subsequently, the hepatic GLP-1 receptor was cascaded to upregulate expression of liver X receptor alpha and ATP-binding cassette transporter A1, thereby reducing pathological VLDL overproduction. CONCLUSION This investigation establishes nuciferine's therapeutic potential in metabolic disorders and hyperlipidemia by activating intestinal TAS2R46 to regulate hepatic VLDL synthesis and secretion.
Collapse
Affiliation(s)
- Chen Ding
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Ministry of Education, Yantai University, Yantai 264005, PR China
| | - Jian Ruan
- Yantai Center for Food and Drug Control, Yantai 264000, PR China
| | - Jingxian Huang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Ministry of Education, Yantai University, Yantai 264005, PR China
| | - Limin Liu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Ministry of Education, Yantai University, Yantai 264005, PR China
| | - Yu Li
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Ministry of Education, Yantai University, Yantai 264005, PR China
| | - Yuan Du
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Ministry of Education, Yantai University, Yantai 264005, PR China.
| | - Yan Zhao
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Ministry of Education, Yantai University, Yantai 264005, PR China.
| |
Collapse
|
8
|
Guilmineau C, Tremblay-Franco M, Vialaneix N, Servien R. Phoenics: a novel statistical approach for longitudinal metabolomic pathway analysis. BMC Bioinformatics 2025; 26:105. [PMID: 40240918 PMCID: PMC12001596 DOI: 10.1186/s12859-025-06118-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 03/20/2025] [Indexed: 04/18/2025] Open
Abstract
BACKGROUND Metabolomics describes the metabolic profile of an organism at a given time by the concentrations of its constituent metabolites. When studied over time, metabolite concentrations can help understand the dynamical evolution of a biological process. However, metabolites are involved into sequences of chemical reactions, called metabolic pathways, related to a given biological function. Accounting for these pathways into statistical methods for metabolomic data is thus a relevant way to directly express results in terms of biological functions and to increase their interpretability. METHODS We propose a new method, phoenics, to perform differential analysis for longitudinal metabolomic data at the pathway level. In short, phoenics proceeds in two steps: First, the matrix of metabolite quantifications is transformed by a dimension reduction approach accounting for pathway information. Then, a mixed linear model is fitted on the transformed data. RESULTS This method was applied to semi-synthetic NMR data and two real NMR datasets assessing the effects of antibiotics and irritable bowel syndrome on feces. Results showed that phoenics properly controls the Type I error rate and has a better ability to detect differential metabolic pathways and to extract new impacted biological functions than alternative methods. The method is implemented in the R package phoenics available on CRAN.
Collapse
Affiliation(s)
- Camille Guilmineau
- INRAE, University of Montpellier, LBE, 102 Avenue des Etangs, 11100, Narbonne, France.
| | - Marie Tremblay-Franco
- INRAE, Université de Toulouse, ENVT, Toxalim, 31027, Toulouse, France
- Axiom Platform, MetaToul-MetaboHUB, National Infrastructure for Metabolomics and Fluxomics, 31027, Toulouse, France
| | | | - Rémi Servien
- INRAE, University of Montpellier, LBE, 102 Avenue des Etangs, 11100, Narbonne, France
| |
Collapse
|
9
|
Peng W, Jin Z, Liu J, Zhang Q, Liu W. Tangeretin modulates gut microbiota metabolism and macrophage immunity following fecal microbiota transplantation in obesity. J Food Sci 2025; 90:e70171. [PMID: 40183701 DOI: 10.1111/1750-3841.70171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 02/25/2025] [Accepted: 03/14/2025] [Indexed: 04/05/2025]
Abstract
Obesity, characterized by excessive body fat, is a leading preventable cause of death globally and represents one of the most critical public health challenges of the 21st century. This study aimed to investigate the action of tangeretin on gut microbiota metabolism and inflammation in high-fat diet (HFD)-induced obese mice. A model of obesity was established using 6-week-old male C57BL/6J mice fed with HFD, which were then used for the treatment with tangeretin (20 mg/kg/mice/day) or antibiotic (Abx). The results showed that the tangeretin intervention alleviated fat deposition and disorder of cellular structural integrity in the model group. The obese mice showed a significant increase in the levels of lipid (glycerol, triglyceride, and total cholesterol), inflammatory factors (IL-6 and TNF-α), and F4/80 expression in both serum and adipose tissues. Following tangeretin treatment, the levels of lipid, inflammatory factors, and the ratio of F4/80 + CD206 + macrophages were decreased in both serum and adipose tissue. 16S rRNA sequencing and LC-MS/MS analysis revealed that tangeretin decreased obesity in HFD-induced obese mice by interacting with gut microbiota, particularly influencing Parabacteroides, Blautia, and Parasutterella, and amino acids such as threonine, isoleucine, leucine, phenylalanine, arginine, glutamine, L-tryptophan, and tyrosine. Abx-mediated clearance of gut microbiota blocked the HFD-induced obesity and abrogated the therapeutic effects of tangeretin in obese mice. Fecal microbiota transplantation (FMT) proved that clearing gut microbiota with Abx blocked the beneficial effects of FMTHFD+Tangeretin intervention. These findings suggested that tangeretin improved HFD-induced obesity by regulating lipid metabolism and modulating F4/80 macrophage activation via gut microbiota.
Collapse
Affiliation(s)
- Weihui Peng
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Zhangliu Jin
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Jinjin Liu
- Department of Biliopancreatic Surgery, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Qirui Zhang
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Wei Liu
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
10
|
Wang G, Wang B, Zhou Q, Cheng Z, Liu L, Zhang S, Zhou S, Luo P. Puerarin combined with Hericium erinaceus insoluble dietary fiber alleviates obesity induced by high-fat diet through regulating the glycerophospholipid metabolism pathway influenced by gut microbiota. Appl Environ Microbiol 2025; 91:e0237624. [PMID: 39976439 PMCID: PMC11921353 DOI: 10.1128/aem.02376-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 01/19/2025] [Indexed: 02/21/2025] Open
Abstract
The purpose of this study was to evaluate the combined effects of puerarin (Pue) and insoluble dietary fiber from Hericium erinaceus (HEIDF) on obesity induced by a high-fat diet (HFD) in mice, focusing on their effects on lipid and glucose metabolism, gut microbiota (GM), and serum metabolites. Glucose tolerance, tissue pathology, and serum biochemical levels were conducted to assess the effects of puerarin combined with Hericium Erinaceus insoluble dietary fiber (LH) on glucose and lipid metabolism. 16S rRNA sequencing and untargeted metabonomics were employed to explore the underlying mechanisms. The results showed that the LH group significantly reduced body weight and hepatic and adipose lipid accumulation, and improved glucose tolerance and dyslipidemia compared to the Pue or HEIDF groups alone. Moreover, the LH group exhibited enhanced regulation of GM, including increased microbial diversity, higher abundance of beneficial bacteria such as g__Lactobacillus and g__Bacillus, and a decreased Firmicutes-to-Bacteroidota ratio. In addition, the LH group ameliorated HFD-induced serum metabolite changes and promoted the activation of tryptophan and glycerophospholipid metabolism pathways. The combination of Pue and HEIDF exhibits a synergistic anti-obesity effect by modulating specific GM (g__Lactobacillus and g__Bacillus) and serum metabolites.IMPORTANCEThe combination of HEIDF and Pue holds significant importance in the context of obesity. This synergistic effect not only aids in weight management but may also enhance metabolic health through various mechanisms, including increased satiety and promotion of fat oxidation. Therefore, incorporating these two components into the daily diet could offer effective strategies for the prevention and intervention of obesity and its related diseases.
Collapse
Affiliation(s)
- Guoze Wang
- The Affiliated Hospital of Guizhou Medical University, Guizhou Provincial Engineering Research Center of Ecological Food Innovation, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang, China
| | - Binbin Wang
- The Affiliated Hospital of Guizhou Medical University, Guizhou Provincial Engineering Research Center of Ecological Food Innovation, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang, China
| | - Qin Zhou
- The Affiliated Hospital of Guizhou Medical University, Guizhou Provincial Engineering Research Center of Ecological Food Innovation, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang, China
| | - Zhimei Cheng
- The Affiliated Hospital of Guizhou Medical University, Guizhou Provincial Engineering Research Center of Ecological Food Innovation, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang, China
| | - Li Liu
- The Affiliated Hospital of Guizhou Medical University, Guizhou Provincial Engineering Research Center of Ecological Food Innovation, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang, China
- The Key Laboratory of Precision Nutrition and Health of Ministry of Education, Harbin Medical University, Harbin, China
| | - Shuai Zhang
- The Affiliated Hospital of Guizhou Medical University, Guizhou Provincial Engineering Research Center of Ecological Food Innovation, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang, China
| | - Shi Zhou
- The Affiliated Hospital of Guizhou Medical University, Guizhou Provincial Engineering Research Center of Ecological Food Innovation, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang, China
| | - Peng Luo
- The Affiliated Hospital of Guizhou Medical University, Guizhou Provincial Engineering Research Center of Ecological Food Innovation, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang, China
| |
Collapse
|
11
|
Yao H, Xu J, Zhou A, Shen D, Dong Q, Yang X, Li M, Mi X, Lu Y, Zhong R, Shi X, Tai Q, Chen G, Shi B, Sun L, Zhou D, Yao Y, He S. Gut Microbiota-Mediated hsa_circ_0126925 Targets BCAA Metabolic Enzyme BCAT2 to Exacerbate Colorectal Cancer Progression. Mol Cancer Res 2025; 23:202-218. [PMID: 39642324 PMCID: PMC11873731 DOI: 10.1158/1541-7786.mcr-24-0434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Accepted: 12/05/2024] [Indexed: 12/08/2024]
Abstract
Recent evidence indicates that a high-fat diet can promote tumor development, especially colorectal cancer, by influencing the microbiota. Regulatory circular RNA (circRNA) plays an important role in modulating host-microbe interactions; however, the specific mechanisms by which circRNAs influence cancer progression by regulating these interactions remain unclear. Here, we report that consumption of a high-fat diet modulates the microbiota by specifically upregulating the expression of the noncoding RNA hsa_circ_0126925 (herein, referred to as circ_0126925) in colorectal cancer. Acting as a scaffold, circ_0126925 hinders the recruitment of the E3 ubiquitin ligase tripartite motif-containing protein 21 (TRIM21) to branched-chain amino acid transaminase 2 (BCAT2), leading to reduced degradation of BCAT2. This reduction in targeted degradation of BCAT2 can protect tumors from limited branched-chain amino acid (BCAA) interference by improving the metabolism of BCAAs in colorectal cancer. Taken together, these data demonstrate that circ_0126925 plays a critical role in promoting the progression of colorectal cancer by maintaining BCAA metabolism and provide insight into the functions and crosstalk of circ_0126925 in host-microbe interactions in colorectal cancer. Implications: This study preliminarily confirms that circRNAs do indeed respond to microbiota/microbial metabolites, providing further evidence for the potential development of circRNAs as diagnostic tools and/or therapeutic agents to alleviate microbiome-related pathology in humans.
Collapse
Affiliation(s)
- Huihui Yao
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jiancheng Xu
- Cancer Institute, Suzhou Medical College, Soochow University, Suzhou, China
| | - Aina Zhou
- Department of Otorhinolaryngology, Head and Neck Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Danyang Shen
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Qiuchen Dong
- Department of Chemistry, Xi’an Jiaotong-Liverpool University, Suzhou, China
| | - Xiaodong Yang
- Department of General Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Mengyu Li
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cam-Su Mouse Genomic Resources Center, Medical College of Soochow University, Suzhou, China
| | - Xiuwei Mi
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yang Lu
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Runze Zhong
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xinyu Shi
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Qingliang Tai
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Guoliang Chen
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Bo Shi
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Liang Sun
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Diyuan Zhou
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yizhou Yao
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Songbing He
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
- Cancer Institute, Suzhou Medical College, Soochow University, Suzhou, China
- Suzhou Biomedical Industry Innovation Center & National Center of Technology Innovation for Biopharmaceuticals, Suzhou, China
| |
Collapse
|
12
|
Li W, Tu J, Zheng J, Das A, Yan Q, Jiang X, Ding W, Bai X, Lai K, Yang S, Yang C, Zou J, Diwan AD, Zheng Z. Gut Microbiome and Metabolome Changes in Chronic Low Back Pain Patients With Vertebral Bone Marrow Lesions. JOR Spine 2025; 8:e70042. [PMID: 39877797 PMCID: PMC11772216 DOI: 10.1002/jsp2.70042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 09/26/2024] [Accepted: 12/13/2024] [Indexed: 01/31/2025] Open
Abstract
Background Chronic low back pain (LBP) is a significant global health concern, often linked to vertebral bone marrow lesions (BML), particularly fatty replacement (FR). This study aims to explore the relationship between the gut microbiome, serum metabolome, and FR in chronic LBP patients. Methods Serum metabolomic profiling and gut microbiome analysis were conducted in chronic LBP patients with and without FR (LBP + FR, n = 40; LBP, n = 40) and Healthy Controls (HC, n = 31). The study investigates alterations in branched-chain amino acids (BCAAs) levels and identifies key microbial species associated with BCAA metabolism. In vitro experiments elucidate the role of BCAAs in adipogenesis of bone marrow mesenchymal stem cells (BM-MSCs) via the SIRT4 pathway. Results Chronic LBP patients with FR exhibit depleted BCAA levels in their serum metabolome, along with alterations in the gut microbiome. Specific microbial species, including Ruminococcus gnavus, Roseburia hominis, and Lachnospiraceae bacterium 8 1 57FAA, are identified as influential in BCAA metabolism and BM-MSCs metabolism. In vitro experiments demonstrate the ability of BCAAs to induce BM-MSCs adipogenesis through SIRT4 pathway activation. Conclusion This study sheds light on the intricate relationship between the disturbed gut ecosystem, serum metabolites, and FR in chronic LBP. Dysbiosis in the gut microbiome may contribute to altered BCAA degradation, subsequently promoting BM-MSCs adipogenesis and FR. Understanding these interactions provides insights for targeted therapeutic strategies to mitigate chronic LBP associated with FR by restoring gut microbial balance and modulating serum metabolite profiles.
Collapse
Affiliation(s)
- Wentian Li
- Spine Labs, St. George and Sutherland Clinical SchoolUniversity of new South WalesKogarahAustralia
- Gulbali Institute, School of Agricultural, Environmental and Veterinary SciencesCharles Sturt UniversityWagga WaggaAustralia
| | - Ji Tu
- Spine Labs, St. George and Sutherland Clinical SchoolUniversity of new South WalesKogarahAustralia
- Nepean HospitalNepean Blue Mountains Local Health DistrictPenrithAustralia
| | - Jinjian Zheng
- Department of Spine Surgery, the First Affiliated HospitalSun Yat‐Sen UniversityGuangzhouChina
| | - Abhirup Das
- Spine Labs, St. George and Sutherland Clinical SchoolUniversity of new South WalesKogarahAustralia
| | - Qi Yan
- Department of Orthopedic SurgeryThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Xiaotao Jiang
- UNSW Microbiome Research Centre, St George and Sutherland Clinical Campuses, School of Clinical Medicine, UNSW Medicine and HealthThe University of new South WalesSydneyAustralia
| | - Wenyuan Ding
- Department of Spinal SurgeryThe Third Hospital of Hebei Medical UniversityShijiazhuangChina
- Hebei Joint International Research Centre for Spinal DiseasesCenter for Innovation & Translational Medicine, the First Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
| | - Xupeng Bai
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Charles Perkins Centre and School of Medical SciencesUniversity of SydneySydneyAustralia
| | - Kaitao Lai
- Northcott Neuroscience LaboratoryANZAC Research Institute, Concord HospitalSydneyAustralia
- Tissue Engineering and Microfluidics Laboratory (TE&M)Australian Institute for Bioengineering and Nanotechnology (AIBN), the University of QueenslandSt LuciaAustralia
| | - Sidong Yang
- Department of Spinal SurgeryThe Third Hospital of Hebei Medical UniversityShijiazhuangChina
- Department of Orthopaedics, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Cao Yang
- Spine ServiceDepartment of Orthopaedic Surgery, St. George HospitalKogarahAustralia
| | - Jun Zou
- Department of Orthopedic SurgeryThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Ashish D. Diwan
- Spine Labs, St. George and Sutherland Clinical SchoolUniversity of new South WalesKogarahAustralia
- Department of Orthopaedics, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Zhaomin Zheng
- Spine Labs, St. George and Sutherland Clinical SchoolUniversity of new South WalesKogarahAustralia
- Department of Spine Surgery, the First Affiliated HospitalSun Yat‐Sen UniversityGuangzhouChina
| |
Collapse
|
13
|
Dai D, Dong C, Kong F, Wang S, Wang S, Wang W, Li S. Dietary supplementation of S cutellariae radix flavonoid extract improves lactation performance in dairy cows by regulating gastrointestinal microbes, antioxidant capacity and immune function. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2025; 20:499-508. [PMID: 40092352 PMCID: PMC11909456 DOI: 10.1016/j.aninu.2024.11.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 08/23/2024] [Accepted: 11/20/2024] [Indexed: 03/19/2025]
Abstract
Scutellariae radix flavonoid extract (SFE) has been acknowledged for its antioxidant, anti-inflammatory and antimicrobial properties in enhancing gastrointestinal microbial communities and improving the host's immunity. Nevertheless, the impacts of dietary supplementation with SFE on the gastrointestinal microbes and host metabolism in dairy cows remain uncertain. Therefore, the aim of this study was to assess the effects of dietary supplementation with SFE on the lactation performance, gastrointestinal microbes, and plasma biochemical parameters of dairy cows. Six ruminally and duodenally cannulated multiparous dairy cows were used in a crossover design over 28-d periods that included a 21-d adaptation and a 7-d sample collection period. Cows were fed a basal diet (CON group) or a basal diet supplemented with SFE at 25 g/d (SFE group). SFE supplementation tended to increase milk yield (P = 0.067) and milk urea N concentration (P = 0.079), and decreased the milk somatic cell counts (SCC, P = 0.036). Cows in the SFE group had lower plasma aspartate aminotransferase (AST), malondialdehyde (MDA), tumor necrosis factor (TNF-α), and interleukin-1β concentrations compared with the CON (P < 0.05). Meanwhile, SFE supplementation increased butyrate concentration in the rumen (P = 0.044). The microbial structure of rumen and duodenum were affected by SFE supplementation (P = 0.009 and P = 0.031; respectively), resulting in enrichment of Butyrivibrio in both parts of the SFE cows (P = 0.034 and P = 0.029; respectively). However, microbial structure and composition of feces were not affected by SFE supplementation. Overall, our study indicated that dietary supplementation with SFE could enhance lactation performance and milk quality in dairy cows by improving the gastrointestinal inner environment and health status.
Collapse
Affiliation(s)
- Dongwen Dai
- State Key Laboratory of Animal Nutrition, Beijing Engineering Technology Research Center of Raw Milk Quality and Safety Control, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
- College of Animal Science and Technology, Ningxia University, Yinchuan 750021, China
| | - Chunxiao Dong
- State Key Laboratory of Animal Nutrition, Beijing Engineering Technology Research Center of Raw Milk Quality and Safety Control, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
- College of Animal Science and Technology, Ningxia University, Yinchuan 750021, China
| | - Fanlin Kong
- State Key Laboratory of Animal Nutrition, Beijing Engineering Technology Research Center of Raw Milk Quality and Safety Control, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Shuo Wang
- State Key Laboratory of Animal Nutrition, Beijing Engineering Technology Research Center of Raw Milk Quality and Safety Control, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | | | - Wei Wang
- State Key Laboratory of Animal Nutrition, Beijing Engineering Technology Research Center of Raw Milk Quality and Safety Control, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Shengli Li
- State Key Laboratory of Animal Nutrition, Beijing Engineering Technology Research Center of Raw Milk Quality and Safety Control, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
- College of Animal Science and Technology, Ningxia University, Yinchuan 750021, China
| |
Collapse
|
14
|
Yang FQ, Tan XM, Chu SS, Yin MZ, Zhang ZY, Peng HS. UPLC-Q-TOF-MS With Chemometrics Approach Analysis of Nonvolatile Compounds for Medicinal Citrus reticulata With Cultivar and Areas Variations. PHYTOCHEMICAL ANALYSIS : PCA 2025; 36:467-484. [PMID: 39731403 DOI: 10.1002/pca.3496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 11/25/2024] [Accepted: 11/27/2024] [Indexed: 12/29/2024]
Abstract
INTRODUCTION Citri Reticulatae Pericarpium (CRP), also known as Chenpi in Chinese, is the dry mature peel of Citrus reticulata Blanco or its cultivated varieties. CRP as the health-care food and dietary supplement has been widely used in various diseases. The quality of CRP can be affected by various factors, which are closely related to the metabolite composition of CRP. OBJECTIVES The objective of this study is to conduct a comprehensive comparative analysis on the chemical profiling of 51 C. reticulata samples of eight medicinal varieties, grown in different areas, and provide a methodological reference for the study of pharmacodynamic material bases and quality control of C. reticulata. METHODOLOGY Initially, a comprehensive characterization was performed using quadrupole time-of-flight mass spectrometry (UPLC-Q-TOF-MS), and a heatmap visualization was employed for clarifying the distribution of the annotated active ingredients. Furthermore, obtained chemical profiles data were employed in multivariate statistical methods, comprising principal component analysis (PCA), and orthogonal partial least-squares-discrimination analysis (OPLS-DA). RESULTS A total of 42 chemical components were annotated in positive ion mode. The relative contents were evident differences in the active ingredients of medicinal varieties of C. reticulata; mostly, polymethoxy flavones (PMFs) in C. reticulata "Dahongpao" were more abundant; among them, nobiletin and tangeretin are the main active ingredients in CRP. In addition, the relative contents of chemical constituents of C. reticulata "Dahongpao" and C. reticulata "Unshiu" from different areas were less variable. Compared with production origins, the varieties of C. reticulata had a greater impact on quality. CONCLUSION This work obtains a better understanding of the chemical profiles of medicinal varieties of C. reticulata, facilitated the reasonable applicability and quality control of medicinal varieties of C. reticulata.
Collapse
Affiliation(s)
- Fang-Qing Yang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiang-Mei Tan
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Shan-Shan Chu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Min-Zhen Yin
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhen-Yu Zhang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Hua-Sheng Peng
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
15
|
Zhu Z, Zuo S, Zhu Z, Wang C, Du Y, Chen F. THSWD upregulates the LTF/AMPK/mTOR/Becn1 axis and promotes lysosomal autophagy in hepatocellular carcinoma cells by regulating gut flora and metabolic reprogramming. Int Immunopharmacol 2025; 148:114091. [PMID: 39826450 DOI: 10.1016/j.intimp.2025.114091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 12/31/2024] [Accepted: 01/11/2025] [Indexed: 01/22/2025]
Abstract
THSWD has the effect of reducing inflammation, improving microcirculation, and regulating immune status in patients with hepatocellular carcinoma. Regardless of its clear therapeutic effect, the underlying mechanism of action against hepatocellular carcinoma is not clear. To identify critical gut microbiota and its associated metabolites related to THSWD inhibition against hepatocellular carcinoma progression, we assessed the microbe-dependent anti-hepatocellular carcinoma effects of THSWD through 16 s rRNA gene sequencing, fecal microbial transplantation and antibiotic treatment. Metabolic analyses, transcriptomic analyses, and molecular experiments were performed to explore how THSWD modulates the gut microbiota against hepatocellular carcinoma progression. As confirmed by in vivo and in vitro assays, THSWD reduced tumour growth rate and promoted apoptosis in hepatocellular carcinoma cells in hepatocellular carcinoma model mice, and liver and kidney indexes were detected and confirmed the safety of THSWD. Transcriptomic analysis revealed that the targets of THSWD were significantly enriched in multiple lysosomal autophagy signalling pathways, suggesting that lysosomal autophagy is probably associated with THSWD's therapeutic effect. Based on the integrated data analysis, THSWD delays hepatocellular carcinoma progression by increasing the intestinal microbiota Duncaniella and augmenting the metabolite glabrol, and the joint analysis of metabolic and genomic data suggests that this metabolite is associated with lysosomal autophagy, and cellular experiments confirmed that the The differential metabolite glabrol induces apoptosis in hepatocellular carcinoma cells by triggering the lysosomal autophagy-mediated apoptosis signalling pathway. Supplementation with glabrol metabolites up regulates the LTF/AMPK/mTOR/Beclin1 axis and promotes hepatocellular carcinoma cells with lysosomal autophagy and induced apoptosis in hepatocellular carcinoma cells.
Collapse
Affiliation(s)
- Zhiqin Zhu
- Department of Hepatology, Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, 510315 Guangzhou, China
| | - Shiqi Zuo
- Department of Pathology, Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou, Guangdong 510315, China
| | - Zhiqi Zhu
- School of Materials Science and Engineering, Central South University, Changsha 410083, China
| | - Chen Wang
- Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Yangfeng Du
- Changde Hospital, Xiangya School of Medicine, Central South University, 415000 Changde, China.
| | - Fengsheng Chen
- Department of Hepatology, Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, 510315 Guangzhou, China.
| |
Collapse
|
16
|
Zhou X, Zhang Y, Wei L, Yang Y, Wang B, Liu C, Bai J, Wang C. In vitro fermentation characteristics of fucoidan and its regulatory effects on human gut microbiota and metabolites. Food Chem 2025; 465:141998. [PMID: 39549519 DOI: 10.1016/j.foodchem.2024.141998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 10/29/2024] [Accepted: 11/08/2024] [Indexed: 11/18/2024]
Abstract
Dietary polysaccharides affect the intestinal microorganisms and their metabolites in the host. Clarifying the relationship among polysaccharides, intestinal microflora, and their metabolites is helpful to formulate dietary nutrition intervention strategies. Thus, we explored the regulatory effects of fucoidan on the human gut microbiota and its metabolites. After 48 h of fermentation, fucoidan significantly reduced the pH value in the broth, accompanied by an increase in total short-chain fatty acids, acetic acid, and propanoic acid contents. Fucoidan significantly reduced the relative abundance of Escherichia_shigella and Blebsiella and increased the relative abundance of Bifidobacterium and Lactobacillus. Concurrently, fucoidan altered the composition of intestinal microbial metabolites. These results indicate that fucoidan can regulate the metabolism of the intestinal flora and host, which may contribute to the intestinal health of the host.
Collapse
Affiliation(s)
- Xu Zhou
- College of Food Science, Southwest University, Chongqing 400715, China
| | - Yuyan Zhang
- College of Food Science, Southwest University, Chongqing 400715, China
| | - Li Wei
- College of Food Science, Southwest University, Chongqing 400715, China
| | - Yuhan Yang
- College of Food Science, Southwest University, Chongqing 400715, China
| | - Botao Wang
- Bloomage Biotechnology CO, LTD, Jinan, Shandong 250000, China
| | - Cuiping Liu
- Department of Radiology, Yuxi Children's Hospital, Yuxi, Yunnan 653100, China
| | - Junying Bai
- Citrus Research Institute, Southwest University, Chongqing 400700, China.
| | - Chen Wang
- College of Food Science, Southwest University, Chongqing 400715, China.
| |
Collapse
|
17
|
Xia Y, Wang L, Qiu Y, Ge W. High-dose thiamine supplementation ameliorates obesity induced by a high-fat and high-fructose diet in mice by reshaping gut microbiota. Front Nutr 2025; 12:1532581. [PMID: 39990607 PMCID: PMC11842239 DOI: 10.3389/fnut.2025.1532581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 01/20/2025] [Indexed: 02/25/2025] Open
Abstract
Introduction Thiamine (vitamin B1) in the gut is crucial for maintaining intestinal homeostasis and host health. Our previous study identified significantly lower levels of fecal thiamine in individuals with obesity; however, its potential and mechanisms for alleviating obesity induced by a high-fat and high-fructose diet (HFFD) remain unclear. Therefore, in the present study, the effects of high-dose thiamine supplementation on HFFD-induced obesity and gut microbiota dysbiosis were investigated. Methods HFFD-fed mice were supplemented with high-dose thiamine for eight weeks. Biochemical analysis and histological analysis were conducted to assess phenotypic changes. Fecal 16S rRNA gene sequencing was performed to analyze alterations in the gut microbiota. Results The results showed that high-dose thiamine supplementation for eight weeks could significantly alleviate symptoms of HFFD-induced obesity and improve HFFD-induced intestinal epithelial barrier dysfunction by enhancing the tight junction function. Furthermore, oral administration of high-dose thiamine also regulated HFFD-induced gut microbiota dysbiosis by reshaping its structure and composition of gut microbiota, such as increasing the relative abundance of Actinobacteria and Bifidobacterium pseudolongum, and reducing the relative abundance of Proteobacteria and Ruminococcus gnavus, accompanied by decreased level of gut-derived endotoxin. Finally, significant correlations were found between obesity-related phenotypes and gut microbiota through correlation analysis. Conclusion Our findings suggest that the potential mechanism by which high-dose thiamine supplementation alleviated HFFD-induced obesity might involve reshaping gut microbiota and restoring the intestinal barrier, thereby ameliorating gut microbiota-related endotoxemia.
Collapse
Affiliation(s)
- Yu Xia
- Department of Pharmacy, China Pharmaceutical University Nanjing Drum Tower Hospital, Nanjing, China
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Lulu Wang
- Department of Pharmacy, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Yanyan Qiu
- Department of Pediatrics, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, China
| | - Weihong Ge
- Department of Pharmacy, China Pharmaceutical University Nanjing Drum Tower Hospital, Nanjing, China
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
- Department of Pharmacy, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| |
Collapse
|
18
|
Fu Y, Wang C, Gao Z, Liao Y, Peng M, Fu F, Li G, Su D, Guo J, Shan Y. Microbes: Drivers of Chenpi manufacturing, biotransformation, and physiological effects. Food Chem 2025; 464:141631. [PMID: 39454433 DOI: 10.1016/j.foodchem.2024.141631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 09/16/2024] [Accepted: 10/11/2024] [Indexed: 10/28/2024]
Abstract
Chenpi holds a rich history of both edible and medicinal applications worldwide, garnering increased attention from researchers in recent years due to its diverse physiological effects. While current research predominantly exploresed its chemical composition and physiological effects, there remains a notable gap in knowledge concerning its manufacturing, characteristic chemical substances, and the underlying mechanisms driving its physiological effects. In this review, the impacts of microbes on the manufacturing, biotransformation, and physiological effects of Chenpi were summarized, as well as the present status of product development. Furthermore, this review engaged in an in-depth discussion highlighting the challenges and shortcomings in recent research, while proposing potential directions and prospects. Additionally, the claim that "The longer the aging, the better the quality" of Chenpi was scientifically evaluated for the first time, providing a solid theoretical foundation for advancing the Chenpi industry.
Collapse
Affiliation(s)
- Yanjiao Fu
- Longping Branch, College of Biology, Hunan University, Changsha 410125, China; Hunan Agriculture Product Processing Institute; Dongting Laboratory; Hunan Academy of Agricultural Sciences, Changsha 410125, China
| | - Chao Wang
- Longping Branch, College of Biology, Hunan University, Changsha 410125, China; Hunan Agriculture Product Processing Institute; Dongting Laboratory; Hunan Academy of Agricultural Sciences, Changsha 410125, China
| | - Zhipeng Gao
- Fisheries College, Hunan Agricultural University, Changsha 410128, China
| | - Yanfang Liao
- Longping Branch, College of Biology, Hunan University, Changsha 410125, China; Hunan Agriculture Product Processing Institute; Dongting Laboratory; Hunan Academy of Agricultural Sciences, Changsha 410125, China
| | - Mingfang Peng
- Longping Branch, College of Biology, Hunan University, Changsha 410125, China; Hunan Agriculture Product Processing Institute; Dongting Laboratory; Hunan Academy of Agricultural Sciences, Changsha 410125, China
| | - Fuhua Fu
- Longping Branch, College of Biology, Hunan University, Changsha 410125, China; Hunan Agriculture Product Processing Institute; Dongting Laboratory; Hunan Academy of Agricultural Sciences, Changsha 410125, China
| | - Gaoyang Li
- Longping Branch, College of Biology, Hunan University, Changsha 410125, China; Hunan Agriculture Product Processing Institute; Dongting Laboratory; Hunan Academy of Agricultural Sciences, Changsha 410125, China
| | - Donglin Su
- Longping Branch, College of Biology, Hunan University, Changsha 410125, China; Hunan Agriculture Product Processing Institute; Dongting Laboratory; Hunan Academy of Agricultural Sciences, Changsha 410125, China
| | - Jiajing Guo
- Hunan Agriculture Product Processing Institute; Dongting Laboratory; Hunan Academy of Agricultural Sciences, Changsha 410125, China.
| | - Yang Shan
- Longping Branch, College of Biology, Hunan University, Changsha 410125, China; Hunan Agriculture Product Processing Institute; Dongting Laboratory; Hunan Academy of Agricultural Sciences, Changsha 410125, China.
| |
Collapse
|
19
|
Zhang Y, Xu D, Cai X, Xing X, Shao X, Yin A, Zhao Y, Wang M, Fan Y, Liu B, Yang H, Zhou W, Li P. Gut Commensal Barnesiella Intestinihominis Ameliorates Hyperglycemia and Liver Metabolic Disorders. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2411181. [PMID: 39741391 PMCID: PMC11848638 DOI: 10.1002/advs.202411181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 12/19/2024] [Indexed: 01/03/2025]
Abstract
Recent studies have highlighted the role of the gut microbiota in type 2 diabetes (T2D). Improving gut microbiota dysbiosis can be a potential strategy for the prevention and management of T2D. Here, this work finds that the abundance of Barnesiella intestinihominis is significantly decreased in the fecal of T2D patients from 2-independent centers. Oral treatment of live B. intestinihominis (LBI) considerably ameliorates hyperglycemia and liver metabolic disorders in HFD/STZ-induced T2D models and db/db mice. LBI-derived acetate has similar protective effects against T2D. Mechanistically, acetate enhances fibroblast growth factor 21 (FGF21) through inhibition of histone deacetylase 9 (HDAC9) to increase H3K27 acetylation at the FGF21 promoter. The screening puerarin from Gegen Qinlian decoction in a gut microbiota-dependent manner improved hyperglycemia and liver metabolic disorders by promoting the growth of B. intestinihominis. This study suggests that gut commensal B. intestinihominis and puerarin, respectively have the potential as a probiotic and prebiotic in the treatment of T2D.
Collapse
Affiliation(s)
- Ye Zhang
- State Key Laboratory of Natural MedicinesChina Pharmaceutical UniversityNanjing211198China
| | - Dong Xu
- State Key Laboratory of Natural MedicinesChina Pharmaceutical UniversityNanjing211198China
| | - Xuyi Cai
- State Key Laboratory of Natural MedicinesChina Pharmaceutical UniversityNanjing211198China
| | - Xue Xing
- State Key Laboratory of Natural MedicinesChina Pharmaceutical UniversityNanjing211198China
| | - Xin Shao
- Nanjing Hospital of Chinese Medicine affiliated to Nanjing University of Chinese MedicineNanjing210022China
| | - Ailing Yin
- Nanjing Hospital of Chinese Medicine affiliated to Nanjing University of Chinese MedicineNanjing210022China
| | - Yanyan Zhao
- Department of EndocrinologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhou450052China
| | - Mengyuan Wang
- State Key Laboratory of Natural MedicinesChina Pharmaceutical UniversityNanjing211198China
| | - Yu‐nuo Fan
- State Key Laboratory of Natural MedicinesChina Pharmaceutical UniversityNanjing211198China
| | - Boao Liu
- State Key Laboratory of Natural MedicinesChina Pharmaceutical UniversityNanjing211198China
| | - Hua Yang
- State Key Laboratory of Natural MedicinesChina Pharmaceutical UniversityNanjing211198China
| | - Wei Zhou
- State Key Laboratory of Natural MedicinesChina Pharmaceutical UniversityNanjing211198China
| | - Ping Li
- State Key Laboratory of Natural MedicinesChina Pharmaceutical UniversityNanjing211198China
| |
Collapse
|
20
|
Chen Z, Zhou J, Zheng X, Xie H, Hao H. Metabolic insights into gut microbiota in the pharmacology of natural medicines. Chin J Nat Med 2025; 23:158-168. [PMID: 39986692 DOI: 10.1016/s1875-5364(25)60820-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 09/12/2024] [Accepted: 10/24/2024] [Indexed: 02/24/2025]
Abstract
Natural medicines (NMs) demonstrate distinct advantages in the clinical management of chronic diseases. Recent years have seen growing recognition of the gut microbiota's role in the efficacy and synergy of NMs, providing new impetus for elucidating the material basis and mechanisms of NMs and their path toward modernization. A fundamental question that has emerged is how NM-microbiota interactions integrate into the multi-target holistic mechanisms of NMs, the answer to which may also illuminate new avenues for drug discovery. Metabolic regulation via small-molecule metabolites has been increasingly implicated in host-microbe interaction. This review presents an integral metabolic perspective on NMs-microbiota interaction in host health and disease. It highlights the emerging understanding of gut microbiota-related metabolic signals implicated in NM components' local and systemic actions. Additionally, it discusses key issues and prospects related to drug development and the translational study of NMs.
Collapse
Affiliation(s)
- Zixin Chen
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; Laboratory of Metabolic Regulation and Drug Target Discovery, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Junchi Zhou
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; Laboratory of Metabolic Regulation and Drug Target Discovery, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Xiao Zheng
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; Laboratory of Metabolic Regulation and Drug Target Discovery, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| | - Hao Xie
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; Laboratory of Metabolic Regulation and Drug Target Discovery, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| | - Haiping Hao
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; Laboratory of Metabolic Regulation and Drug Target Discovery, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
21
|
Mansoori S, Ho MY, Ng KK, Cheng KK. Branched-chain amino acid metabolism: Pathophysiological mechanism and therapeutic intervention in metabolic diseases. Obes Rev 2025; 26:e13856. [PMID: 39455059 PMCID: PMC11711082 DOI: 10.1111/obr.13856] [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: 11/24/2023] [Revised: 09/02/2024] [Accepted: 10/01/2024] [Indexed: 10/28/2024]
Abstract
Branched-chain amino acids (BCAAs), including leucine, isoleucine, and valine, are essential for maintaining physiological functions and metabolic homeostasis. However, chronic elevation of BCAAs causes metabolic diseases such as obesity, type 2 diabetes (T2D), and metabolic-associated fatty liver disease (MAFLD). Adipose tissue, skeletal muscle, and the liver are the three major metabolic tissues not only responsible for controlling glucose, lipid, and energy balance but also for maintaining BCAA homeostasis. Under obese and diabetic conditions, different pathogenic factors like pro-inflammatory cytokines, lipotoxicity, and reduction of adiponectin and peroxisome proliferator-activated receptors γ (PPARγ) disrupt BCAA metabolism, leading to excessive accumulation of BCAAs and their downstream metabolites in metabolic tissues and circulation. Mechanistically, BCAAs and/or their downstream metabolites, such as branched-chain ketoacids (BCKAs) and 3-hydroxyisobutyrate (3-HIB), impair insulin signaling, inhibit adipogenesis, induce inflammatory responses, and cause lipotoxicity in the metabolic tissues, resulting in multiple metabolic disorders. In this review, we summarize the latest studies on the metabolic regulation of BCAA homeostasis by the three major metabolic tissues-adipose tissue, skeletal muscle, and liver-and how dysregulated BCAA metabolism affects glucose, lipid, and energy balance in these active metabolic tissues. We also summarize therapeutic approaches to restore normal BCAA metabolism as a treatment for metabolic diseases.
Collapse
Affiliation(s)
- Shama Mansoori
- Department of Health Technology and InformaticsThe Hong Kong Polytechnic UniversityHong Kong, China
| | - Melody Yuen‐man Ho
- Department of Health Technology and InformaticsThe Hong Kong Polytechnic UniversityHong Kong, China
| | - Kelvin Kwun‐wang Ng
- Department of Health Technology and InformaticsThe Hong Kong Polytechnic UniversityHong Kong, China
| | - Kenneth King‐yip Cheng
- Department of Health Technology and InformaticsThe Hong Kong Polytechnic UniversityHong Kong, China
- Hong Kong Polytechnic University Shenzhen Research InstituteShenzhenChina
| |
Collapse
|
22
|
Yang M, Xie Q, Wang J, Zha A, Chen J, Jiang Q, Kang M, Deng Q, Yin Y, Tan B. Ningxiang pig-derived lactobacillus reuteri modulates host intramuscular fat deposition via branched-chain amino acid metabolism. MICROBIOME 2025; 13:32. [PMID: 39891238 PMCID: PMC11786426 DOI: 10.1186/s40168-024-02013-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 12/17/2024] [Indexed: 02/03/2025]
Abstract
BACKGROUND Gut microbiota has been extensively demonstrated to modulate host lipid metabolism. Higher intramuscular fat (IMF) accumulation in Chinese indigenous breed pigs is associated with their special gut microbiota structure. However, the specific microbes and metabolic pathways responsible for lipid deposition are still poorly understood. RESULTS In the present study, a comparative analysis of the gut microbiota and metabolome in obese Ningxiang (NX) pigs and lean Duroc × Landrace × Yorkshire (DLY) pigs was conducted. The results revealed a higher abundance of gut lactobacilli and a correlation of branched-chain amino acid (BCAA) metabolism pathway in NX pigs. We proceeded to verify the roles of various lactobacilli strains originating from NX pigs in BCAA metabolism and lipids deposition in SD rats. We demonstrated that L. reuteri is a fundamental species responsible for modulating lipid deposition in NX pigs and that increased circulating levels of BCAA are positively linked to greater lipid deposition. Additionally, it has been verified that L. reuteri originating from NX pigs has the ability to improve BCAA synthesis in the gut and enhance IMF content in lean DLY pigs. The expression of genes related to lipid synthesis was also significantly upregulated. CONCLUSIONS Taken together, our results imply that NX pig-derived L. reuteri regulates BCAA metabolism and plays a potential role in improving the meat quality of lean pig breeds through modulation of host intramuscular lipid deposition. The results provide a new strategy for improving the meat quality of commercial pigs by influencing host metabolism through supplementing dietary additives. Video Abstract.
Collapse
Affiliation(s)
- Mei Yang
- Hunan Provincial Key Laboratory for the Products Quality Regulation of Livestock and Poultry, College of Animal Science and Technology, Hunan Agricultural University, Changsha, Hunan, 410128, P. R. China
- Yuelushan Laboratory, Changsha, Hunan, 410128, P. R. China
| | - Qian Xie
- Hunan Provincial Key Laboratory for the Products Quality Regulation of Livestock and Poultry, College of Animal Science and Technology, Hunan Agricultural University, Changsha, Hunan, 410128, P. R. China
- Yuelushan Laboratory, Changsha, Hunan, 410128, P. R. China
| | - Jing Wang
- Hunan Provincial Key Laboratory for the Products Quality Regulation of Livestock and Poultry, College of Animal Science and Technology, Hunan Agricultural University, Changsha, Hunan, 410128, P. R. China
- Yuelushan Laboratory, Changsha, Hunan, 410128, P. R. China
| | - Andong Zha
- Yuelushan Laboratory, Changsha, Hunan, 410128, P. R. China
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, 410125, P. R. China
| | - Jiashun Chen
- Hunan Provincial Key Laboratory for the Products Quality Regulation of Livestock and Poultry, College of Animal Science and Technology, Hunan Agricultural University, Changsha, Hunan, 410128, P. R. China
- Yuelushan Laboratory, Changsha, Hunan, 410128, P. R. China
| | - Qian Jiang
- Hunan Provincial Key Laboratory for the Products Quality Regulation of Livestock and Poultry, College of Animal Science and Technology, Hunan Agricultural University, Changsha, Hunan, 410128, P. R. China
- Yuelushan Laboratory, Changsha, Hunan, 410128, P. R. China
| | - Meng Kang
- Hunan Provincial Key Laboratory for the Products Quality Regulation of Livestock and Poultry, College of Animal Science and Technology, Hunan Agricultural University, Changsha, Hunan, 410128, P. R. China
- Yuelushan Laboratory, Changsha, Hunan, 410128, P. R. China
| | - Qiuchun Deng
- Hunan Provincial Key Laboratory for the Products Quality Regulation of Livestock and Poultry, College of Animal Science and Technology, Hunan Agricultural University, Changsha, Hunan, 410128, P. R. China
- Yuelushan Laboratory, Changsha, Hunan, 410128, P. R. China
| | - Yulong Yin
- Yuelushan Laboratory, Changsha, Hunan, 410128, P. R. China.
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, 410125, P. R. China.
| | - Bie Tan
- Hunan Provincial Key Laboratory for the Products Quality Regulation of Livestock and Poultry, College of Animal Science and Technology, Hunan Agricultural University, Changsha, Hunan, 410128, P. R. China.
- Yuelushan Laboratory, Changsha, Hunan, 410128, P. R. China.
| |
Collapse
|
23
|
Liu C, Du J, Yang J, Li J, Zhou T, Yu J, Wang X, Lin J, Liang Y, Shi R, Luo R, Shen X, Wang Y, Zhang L, Shu Z. Research on the mechanism of buyang huanwu decoction in the amelioration of age-associated memory impairment based on the "co-occurrence network regulation of intestinal microecology-host metabolism-immune function". JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118819. [PMID: 39303964 DOI: 10.1016/j.jep.2024.118819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 09/07/2024] [Accepted: 09/09/2024] [Indexed: 09/22/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Brain aging can promote neuronal damage, contributing to aging-related diseases like memory dysfunction. Buyang Huanwu Decoction (BYHWD), a traditional Chinese medicine formula known for tonifying qi and activating blood circulation, shows neuroprotective properties. Despite this, the specific mechanism by which BYHWD improves age-associated memory impairment (AAMI) has not been explored in existing literature. AIM OF THE STUDY This study aimed to investigate the mechanism of BYHWD in the improvement of AAMI based on the "co-occurrence network regulation of intestinal microecology-host metabolism-immune function". MATERIALS AND METHODS Firstly, D-galactose was performed to induce a rat model of AAMI. Learning and memory deficits was assessed by the Morris water maze test. H&E and Nissl staining were used to observe the pathological changes in neurons in the hippocampus of rats. Meanwhile, the levels of pro-inflammatory cytokines and the activation of antioxidant enzymes in rat serum were measured using ELISA. Finally, an integrated pharmacological approach was applied to explore the potential mechanism of BYHWD in improving AAMI. RESULTS Our results indicated that BYHWD significantly mitigated the pathological structure of the hippocampus, reversed the levels of IL-6, TNF-α, GSH, and CAT in the serum, and improved learning and memory in aging rats. Transcriptomics combined with network pharmacology showed that energy metabolism and the inflammatory response were the key biological pathways for BYHWD to ameliorate AAMI. Integrative analysis of the microbiome and metabolomics revealed that BYHWD has the potential to restore the balance of abundance between probiotics and harmful bacteria, and ameliorate the reprogramming of energy metabolism caused by aging in the brain. The co-occurrence network analysis demonstrated that a strong correlation between the treatment of AAMI and the stability of intestinal microecology, host metabolism, and immune network. CONCLUSION The findings of this study collectively support the notion that BYHWD has a superior therapeutic effect in an AAMI rat model. The mechanism involves regulating the "intestinal microecology-metabolism-immune function co-occurrence network" system to restore the composition of gut microbiota and metabolites. This further improves the metabolic phenotype of brain tissue and maintains the homeostasis of central nervous system's immunity, leading to an improvement in AAMI. Consequently, this study offers a unique perspective on the prevention and treatment of AAMI. And, BYHWD is also considered to be a promising preclinical treatment for improving AAMI.
Collapse
Affiliation(s)
- Caiyan Liu
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Department of Biology, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai, 519087, China; School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Jieyong Du
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Department of Biology, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai, 519087, China; School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, China; Department of Cardiology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510080, China
| | - Ji Yang
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Jianhua Li
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Department of Biology, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai, 519087, China; School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Tong Zhou
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Department of Biology, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai, 519087, China; School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Jiaming Yu
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Department of Biology, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai, 519087, China; School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Xiao Wang
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Department of Biology, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai, 519087, China; School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Jiazi Lin
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Department of Biology, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai, 519087, China; School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Yefang Liang
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Department of Biology, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai, 519087, China; School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Ruixiang Shi
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Department of Biology, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai, 519087, China; School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Rongfeng Luo
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Department of Biology, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai, 519087, China; School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Xuejuan Shen
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Department of Biology, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai, 519087, China; School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, China; Department of Cardiology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510080, China
| | - Yi Wang
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, China.
| | - Li Zhang
- Department of Cardiology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510080, China.
| | - Zunpeng Shu
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Department of Biology, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai, 519087, China.
| |
Collapse
|
24
|
Sun M, Liang J, Peng Y, Qin L, Ma D, Cai X, Ran L, Wang Y, Wang H, Yang C, Liu X, Deng Z. Enrichment of polymethoxyflavones from citrus fruits using an optimized enzyme/acid-catalyzed hybrid hydrolysis process and its influence on mice gut microbiota. Food Funct 2025; 16:510-523. [PMID: 39679913 DOI: 10.1039/d4fo03391d] [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: 12/17/2024]
Abstract
Citrus polymethoxyflavones (PMFs) have considerable medicinal, health-promoting, and commercial importance. To provide a stable and reliable source of PMFs, an efficient process of large-scale preparation is warranted. Here, an extraction model for enriching PMFs from citrus fruits was proposed using an enzyme/acid-catalyzed hybrid hydrolysis approach. This method was optimized using response surface methodology (RSM). Furthermore, this model was applied to ten citrus varieties to prepare PMF-rich extracts, and six main PMFs were qualitatively and quantitatively analyzed using UPLC-ESI-MS/MS. Among the ten investigated citrus extracts, nobiletin was the most predominant PMF. The total yields of the six PMFs were ranked as C. unshiu > C. reticulata > C. sinensis, indicating that C. unshiu was the most suitable raw material for PMF preparation. Additionally, the PMF-rich extracts showed beneficial regulatory effects on gut microbiota, highlighting their potential health-promoting and therapeutic functions, which warrant further exploration.
Collapse
Affiliation(s)
- Meng Sun
- Hubei Key Laboratory of Natural Products Research and Development, College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, China.
- Key Laboratory of Functional Yeast, China National Light Industry, College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, China
| | - Jianjia Liang
- Hubei Key Laboratory of Natural Products Research and Development, College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, China.
- Key Laboratory of Functional Yeast, China National Light Industry, College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, China
| | - Yan Peng
- The First College of Clinical Medical Science, China Three Gorges University & Yichang Central People's Hospital, Yichang 443002, China
| | - Leilei Qin
- Three Gorges Public Inspection and Testing Center, Yichang, 443005, China
| | - Dongxu Ma
- Hubei Key Laboratory of Natural Products Research and Development, College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, China.
- Key Laboratory of Functional Yeast, China National Light Industry, College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, China
| | - Xiaorong Cai
- Three Gorges Public Inspection and Testing Center, Yichang, 443005, China
| | - Lu Ran
- Hubei Key Laboratory of Natural Products Research and Development, College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, China.
- Key Laboratory of Functional Yeast, China National Light Industry, College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, China
| | - Yueyi Wang
- Hubei Key Laboratory of Natural Products Research and Development, College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, China.
- Key Laboratory of Functional Yeast, China National Light Industry, College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, China
| | - Huimin Wang
- Hubei Key Laboratory of Natural Products Research and Development, College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, China.
- Key Laboratory of Functional Yeast, China National Light Industry, College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, China
| | - Changying Yang
- Hubei Key Laboratory of Natural Products Research and Development, College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, China.
- Key Laboratory of Functional Yeast, China National Light Industry, College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, China
| | - Xiaowen Liu
- Hubei Key Laboratory of Natural Products Research and Development, College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, China.
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Science, China Three Gorges University, Yichang 443002, China
| | - Zhangshuang Deng
- Hubei Key Laboratory of Natural Products Research and Development, College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, China.
- Key Laboratory of Functional Yeast, China National Light Industry, College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, China
| |
Collapse
|
25
|
Li X, Xie E, Sun S, Shen J, Ding Y, Wang J, Peng X, Zheng R, Farag MA, Xiao J. Flavonoids for gastrointestinal tract local and associated systemic effects: A review of clinical trials and future perspectives. J Adv Res 2025:S2090-1232(25)00033-5. [PMID: 39798849 DOI: 10.1016/j.jare.2025.01.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 01/06/2025] [Accepted: 01/06/2025] [Indexed: 01/15/2025] Open
Abstract
BACKGROUND Flavonoids are naturally occurring dietary phytochemicals with significant antioxidant effects aside from several health benefits. People often consume them in combination with other food components. Compiling data establishes a link between bioactive flavonoids and prevention of several diseases in animal models, including cardiovascular diseases, diabetes, gut dysbiosis, and metabolic dysfunction-associated steatotic liver disease (MASLD). However, numerous clinical studies have demonstrated the ineffectiveness of flavonoids contradicting rodent models, thereby challenging the validity of using flavonoids as dietary supplements. AIM OF REVIEW This review provides a clinical perspective to emphasize the effective roles of dietary flavonoids as well as to summarize their specific mechanisms in animals briefly. KEY SCIENTIFIC CONCEPTS OF REVIEW First, this review offers an in-depth elucidation of the metabolic processes of flavonoids within human, encompassing the small, large intestine, and the liver. Furthermore, the review provides a comprehensive overview of the various functions of flavonoids in the gastrointestinal tract, including hindering the breakdown and assimilation of macronutrients, such as polysaccharides and lipids, regulating gut hormone secretion as well as inhibition of mineral iron absorption. In the large intestine, an unabsorbed major portion of flavonoids interact with the gut flora leading to their biotransformation. Once absorbed and circulated in the bloodstream, bioactive flavonoids or their metabolites exert numerous beneficial systemic effects. Lastly, we examine the protective effects of flavonoids in several metabolic disorders, including endothelial dysfunction, MASLD, cardiovascular disease, obesity, hyperlipidemia, and insulin resistance. In conclusion, this review outlines the safety and future prospects of flavonoids in the field of health, especially in the prevention of metabolic syndrome (MetS).
Collapse
Affiliation(s)
- Xiaopeng Li
- Center of Nutrition and Food Sciences Hunan Agricultural Products Processing Institute Hunan Academy of Agricultural Sciences Changsha China.
| | - Enjun Xie
- School of Public Health Zhejiang University School of Medicine Hangzhou China
| | - Shumin Sun
- School of Public Health Zhejiang University School of Medicine Hangzhou China
| | - Jie Shen
- School of Public Health Zhejiang University School of Medicine Hangzhou China
| | - Yujin Ding
- National Clinical Research Center for Metabolic Diseases Metabolic Syndrome Research Center Department of Metabolism and Endocrinology The Second Xiangya Hospital of Central South University Changsha China
| | - Jiaqi Wang
- Ausnutria Dairy Co., Ltd., Changsha 410200 China
| | - Xiaoyu Peng
- Ausnutria Dairy Co., Ltd., Changsha 410200 China
| | - Ruting Zheng
- Ausnutria Dairy Co., Ltd., Changsha 410200 China
| | - Mohamed A Farag
- Pharmacognosy Department, Faculty of Pharmacy, Cairo University, Cairo 11562 Egypt
| | - Jianbo Xiao
- Universidade de Vigo, Nutrition and Bromatology Group, Department of Analytical Chemistry and Food Science, Instituto de Agroecoloxía e Alimentación (IAA) - CITEXVI 36310 Vigo, Spain; Research Group on Food, Nutritional Biochemistry and Health, Universidad Europea del Atlántico, Isabel Torres 21 39011 Santander, Spain.
| |
Collapse
|
26
|
Wang C, Peng M, Gao Z, Fu F, Li G, Su D, Huang L, Guo J, Shan Y. Citrus aurantium 'Changshan-huyou' physiological premature fruit drop: A promising prebiotic to tackle obesity. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 136:156347. [PMID: 39765038 DOI: 10.1016/j.phymed.2024.156347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 09/22/2024] [Accepted: 12/24/2024] [Indexed: 01/16/2025]
Abstract
BACKGROUND Presently, the mitigation and governance of obesity have surfaced as significant public health dilemmas on a global scale. A wealth of studies indicated that the host gut microbiota is instrumental in regulating the interplay between high-fat diet (HFD) intake and the pathogenesis of obesity. Physiological premature fruit drop, a major byproduct of citrus, is rich in a variety of bioactive constituents, yet its potential has remained underutilized for an extended period. PURPOSE The objective of this investigation is to examine the chemical constituents of Citrus aurantium'Changshan-huyou' premature fruit drop (HYFD) and investigate its anti-obesity effects, elucidating its potential pathways. METHODS Volatile compounds and flavonoids in HYFD were analyzed using chromatographic and mass spectrometric techniques. Furthermore, this study utilized biochemical assays and histopathological examinations to evaluate the effects of HYFD on HFD-fed mice. The impact of HYFD on the gut microbiota of the mice was examined through 16S rRNA gene sequencing, and fecal microbiota transplantation was employed to validate the role of the gut microbial community in host obesity prevention. Concurrently, transcriptome was employed to identify differentially expressed genes, providing further insights into the molecular mechanisms through which HYFD manifests its anti-obesity effects. RESULTS Our findings demonstrated that HYFD supplementation significantly alleviated adiposity and ameliorated the dysbiosis of gut microbiota in HFD-induced mice. HYFD rectified the HFD-induced gut microbiota dysregulation, enhanced the presence of beneficial microbial taxa linked to lipid metabolism, including Parabacteroides and Alistipes, and elevated concentrations of the anti-obesity short-chain fatty acids, comprising caproic acid and isocaproic acid. Additionally, transcriptomic analyses confirmed that HYFD prevented obesity in mice by enhancing fatty acid catabolism via the activation of the AMPK/PPARα/CPT1a signaling pathway. CONCLUSION Our results provided novel insights into the mechanism of citrus physiological premature fruit drop and its potential role in preventing obesity, while sparking greater interest in leveraging more biomass waste.
Collapse
Affiliation(s)
- Chao Wang
- Hunan Agriculture Product Processing Institute; Dongting Laboratory; Hunan Academy of Agricultural Sciences, Changsha, Hunan Province 410125, China
| | - Mingfang Peng
- Hunan Agriculture Product Processing Institute; Dongting Laboratory; Hunan Academy of Agricultural Sciences, Changsha, Hunan Province 410125, China
| | - Zhipeng Gao
- Fisheries College, Hunan Agricultural University, Changsha, Hunan Province 410128, China
| | - Fuhua Fu
- Hunan Agriculture Product Processing Institute; Dongting Laboratory; Hunan Academy of Agricultural Sciences, Changsha, Hunan Province 410125, China
| | - Gaoyang Li
- Hunan Agriculture Product Processing Institute; Dongting Laboratory; Hunan Academy of Agricultural Sciences, Changsha, Hunan Province 410125, China
| | - Donglin Su
- Hunan Agriculture Product Processing Institute; Dongting Laboratory; Hunan Academy of Agricultural Sciences, Changsha, Hunan Province 410125, China
| | - Lvhong Huang
- Hunan Agriculture Product Processing Institute; Dongting Laboratory; Hunan Academy of Agricultural Sciences, Changsha, Hunan Province 410125, China
| | - Jiajing Guo
- Hunan Agriculture Product Processing Institute; Dongting Laboratory; Hunan Academy of Agricultural Sciences, Changsha, Hunan Province 410125, China.
| | - Yang Shan
- Hunan Agriculture Product Processing Institute; Dongting Laboratory; Hunan Academy of Agricultural Sciences, Changsha, Hunan Province 410125, China.
| |
Collapse
|
27
|
Ye H, Wang H, Han B, Chen K, Wang X, Ma F, Cheng L, Zheng S, Zhao X, Zhu J, Li J, Hong M. Guizhi Shaoyao Zhimu decoction inhibits neutrophil extracellular traps formation to relieve rheumatoid arthritis via gut microbial outer membrane vesicles. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 136:156254. [PMID: 39586125 DOI: 10.1016/j.phymed.2024.156254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 11/01/2024] [Accepted: 11/11/2024] [Indexed: 11/27/2024]
Abstract
BACKGROUND Rheumatoid arthritis (RA) is a common autoimmune disease with a high disability rate. Accumulating studies suggest that neutrophil extracellular traps (NETs) play a crucial role in the pathogenesis of RA and targeting NETs has emerged as a potential therapeutic strategy for RA. As a traditional Chinese medicine, Guizhi-Shaoyao-Zhimu Decoction (GSZD) has exhibited good efficacy in the treatment of rheumatoid arthritis (RA), while the underly mechanism especially the possibility that GSZD alter NETs formation to relieve RA remains unknown. PURPOSE Our study aimed to investigate relationship between GSZD and NETs in RA treatment and revealed underlying mechanism. METHODS We constructed collagen-induced arthritis (CIA) model and treated CIA mice with GZSY to validate therapeutic effects of GSZD and examine whether GZSD could inhibit NETs formation in RA. And 16S rRNA sequencing and Fecal microbiota transplantation (FMT) experiment were performed to determine whether GSZD could reduce NETs formation to alleviate RA in gut microbiota-associated manner and identify crucial bacterium in response to GSZD administration. CIA mice treated with effective bacteria and its outer membrane vesicles (OMVs) with oral administration to investigate protective effect against RA and NETs regulative efficiency. We utilized small interfering RNA in vivo and vitro to silence gene mediating effect of GZSD-gut microbiota-NETs. RESULTS GSZD could inhibit NETs formation and relive arthritis in CIA mice. Additionally, GSZD alter gut microbiota composition and significantly increase intestinal Parabacteroides goldsteinii (P.goldsteinii) abundance. Mechanistically, P.goldsteinii enriched by GSZD secreted outer membrane vesicles (OMVs) to translocate into joints and activate Cav-1-Nrf2 axis, leading to reduced NETs formation and alleviate arthritis. In clinical, the abundance of P.goldsteinii exhibited negative correlation with NETs indexes and RA disease activities. CONCLUSION Our findings suggest that GSZD inhibits NETs formation to relieve RA in P.goldsteinii-Cav-1-Nrf2 associated manner, which could provide new sight of the prevention and treatment of RA.
Collapse
Affiliation(s)
- Haixin Ye
- Department of Rheumatology and Immunology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Traditional Chinese Internal Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Hao Wang
- Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Bingqi Han
- Department of Rheumatology and Immunology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Traditional Chinese Internal Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Keshan Chen
- Department of Rheumatology and Immunology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Traditional Chinese Internal Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Xing Wang
- Department of Rheumatology and Immunology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Fopei Ma
- Department of Rheumatology and Immunology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Traditional Chinese Internal Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Lifang Cheng
- Department of Rheumatology and Immunology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Songyuan Zheng
- Department of Rheumatology and Immunology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xueqin Zhao
- Department of Traditional Chinese Internal Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Junqing Zhu
- Department of Rheumatology and Immunology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Juan Li
- Department of Rheumatology and Immunology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Traditional Chinese Internal Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China.
| | - Mukeng Hong
- Department of Rheumatology and Immunology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
28
|
Chen X, Sun W, Ji S, Liu X, Hu Y, Zhou X, Zhou B, Ren J, Li B, Liang H. Citrus Polymethoxyflavones Regulate against Aging-Associated Diseases: Advances in Biological Mechanisms Responsible for Their Regulation. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:28209-28224. [PMID: 39661568 DOI: 10.1021/acs.jafc.4c08004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2024]
Abstract
As the proportion of the aging population globally is surging year by year, age-associated diseases, including neurodegenerative, metabolic, and cardiovascular diseases, have recently attracted widespread attention of food scientists and nutritionists. Polymethoxyflavonoids (PMFs), a type of dietary flavonoids, have emerged as potential antiaging candidates owing to their diverse bioactivities, encompassing antioxidant, anti-inflammatory, neuroprotective, and metabolic regulatory effects. Herein, this comprehensive updated review has summarized and discussed the effects of PMFs on aging, and the possible mechanisms that link PMFs-mediated modulation and the prevention or treatment of various aging-related diseases have been elaborated in detail. Furthermore, the biological fate of PMFs have been discussed elaborately from their absorption, distribution, metabolism, and excretion in vivo. Special attention is given to the bioavailability-bioactivity relationship of PMFs, as PMF's biological activity is significantly hampered by poor bioavailability. Overall, all of these conclusions may help in providing a perspective for further study of PMFs on aging.
Collapse
Affiliation(s)
- Xiaojuan Chen
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
- Key Laboratory of Environment Correlative Dietology (Huazhong Agricultural University), Ministry of Education, Wuhan 430070, China
| | - Weiyi Sun
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Sicheng Ji
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Xiang Liu
- Wuhan Senlan Biotechnology Co., Ltd, Wuhan 430120, China
| | - Yueqi Hu
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Xinyue Zhou
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Bin Zhou
- Key Laboratory of Fermentation Engineering, Ministry of Education, National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei Key Laboratory of Industrial Microbiology, School of Biological Engineering and Food, Hubei University of Technology, Wuhan 430068, China
| | - Jingnan Ren
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
- Key Laboratory of Environment Correlative Dietology (Huazhong Agricultural University), Ministry of Education, Wuhan 430070, China
| | - Bin Li
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
- Key Laboratory of Environment Correlative Dietology (Huazhong Agricultural University), Ministry of Education, Wuhan 430070, China
- Wuhan Senlan Biotechnology Co., Ltd, Wuhan 430120, China
| | - Hongshan Liang
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
- Key Laboratory of Environment Correlative Dietology (Huazhong Agricultural University), Ministry of Education, Wuhan 430070, China
| |
Collapse
|
29
|
Cai H, Wang Q, Han X, Zhang H, Wang N, Huang Y, Yang P, Zhang R, Meng K. In Vitro Evaluation of Probiotic Activities and Anti-Obesity Effects of Enterococcus faecalis EF-1 in Mice Fed a High-Fat Diet. Foods 2024; 13:4095. [PMID: 39767037 PMCID: PMC11675756 DOI: 10.3390/foods13244095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 12/14/2024] [Accepted: 12/15/2024] [Indexed: 01/11/2025] Open
Abstract
This research sought to assess the anti-obesity potential of Enterococcus faecalis EF-1. An extensive and robust in vitro methodology confirmed EF-1's significant potential in combating obesity, probably due to its excellent gastrointestinal tract adaptability, cholesterol-lowering property, bile salt hydrolase activity, α-glucosidase inhibition, and fatty acid absorption ability. Moreover, EF-1 exhibited antimicrobial activity against several pathogenic strains, lacked hemolytic activity, and was sensitive to all antibiotics tested. To further investigate EF-1's anti-obesity properties in vivo, a high-fat diet (HFD) was used to induce obesity in C57BL/6J mice. Treatment with EF-1 (2 × 109 CFU/day) mitigated HFD-induced body weight gain, reduced adipose tissue weight, and preserved liver function. EF-1 also ameliorated obesity-associated microbiota imbalances, such as decreasing the Firmicutes/Bacteroidetes ratio and boosting the levels of bacteria (Faecalibacterium, Mucispirillum, Desulfovibrio, Bacteroides, and Lachnospiraceae_NK4A136_group), which are responsible for the generation of short-chain fatty acids (SCFAs). Concurrently, the levels of total SCFAs were elevated. Thus, following comprehensive safety and efficacy assessments in vitro and in vivo, our results demonstrate that E. faecalis EF-1 inhibits HFD-induced obesity through the regulation of gut microbiota and enhancing SCFA production. This strain appears to be a highly promising candidate for anti-obesity therapeutics or functional foods.
Collapse
Affiliation(s)
- Hongying Cai
- Key Laboratory of Feed Biotechnology of Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (H.C.); (H.Z.); (Y.H.); (P.Y.)
| | - Qingya Wang
- Key Laboratory of Yunnan for Biomass Energy and Biotechnology of Environment, Yunnan Normal University, Kunming 650500, China; (Q.W.); (X.H.); (N.W.)
| | - Xiling Han
- Key Laboratory of Yunnan for Biomass Energy and Biotechnology of Environment, Yunnan Normal University, Kunming 650500, China; (Q.W.); (X.H.); (N.W.)
| | - Haiou Zhang
- Key Laboratory of Feed Biotechnology of Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (H.C.); (H.Z.); (Y.H.); (P.Y.)
| | - Na Wang
- Key Laboratory of Yunnan for Biomass Energy and Biotechnology of Environment, Yunnan Normal University, Kunming 650500, China; (Q.W.); (X.H.); (N.W.)
| | - Yuyin Huang
- Key Laboratory of Feed Biotechnology of Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (H.C.); (H.Z.); (Y.H.); (P.Y.)
| | - Peilong Yang
- Key Laboratory of Feed Biotechnology of Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (H.C.); (H.Z.); (Y.H.); (P.Y.)
| | - Rui Zhang
- Key Laboratory of Yunnan for Biomass Energy and Biotechnology of Environment, Yunnan Normal University, Kunming 650500, China; (Q.W.); (X.H.); (N.W.)
- Key Laboratory of Yunnan Provincial Education Department for Plateau Characteristic Food Enzymes, Yunnan Normal University, Kunming 650500, China
| | - Kun Meng
- Key Laboratory of Feed Biotechnology of Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (H.C.); (H.Z.); (Y.H.); (P.Y.)
| |
Collapse
|
30
|
Felicianna, Lo EKK, Chen C, Ismaiah MJ, Zhang F, Leung HKM, El-Nezami H. Low-dose valine attenuates diet-induced metabolic dysfunction-associated steatotic liver disease (MASLD) in mice by enhancing leptin sensitivity and modulating the gut microbiome. Mol Metab 2024; 90:102059. [PMID: 39489290 PMCID: PMC11616088 DOI: 10.1016/j.molmet.2024.102059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 10/10/2024] [Accepted: 10/25/2024] [Indexed: 11/05/2024] Open
Abstract
OBJECTIVES Elevated circulating branched-chain amino acids (BCAAs) have been associated with obesity, insulin resistance, and MASLD. Nonetheless, BCAA supplementation has been shown to provide protective outcomes towards the intervention of MASLD. Currently, there is a lack of study towards the contribution of the BCAA: valine on MASLD. Herein, the effect of low-dose valine supplementation was investigated for its role in the progression of MASLD. METHODS C57BL/6J mice were fed a high-fat/high-cholesterol diet (HFD) to induce MASLD. Upon the establishment of MASLD, valine was supplemented via voluntary oral administration. Clinical and biochemical parameters associated with MASLD were measured, and molecular mechanism and gut microbiota modulation from the effect of valine were investigated. RESULTS Low-dose valine was found to attenuate the progression of MASLD, significantly reducing the gain in body weight, liver weight, and epididymal white adipose tissue (eWAT) weight, while also attenuating hyperglycemia and hyperleptinemia, and improving serum lipid profiles. Mechanistically, in the liver, genes related to hepatic lipogenesis and cholesterol biosynthesis were downregulated, while those associated with fatty acid oxidation, autophagy, and antioxidant capacity were upregulated, and AMPK pathway activity was enhanced. Liver and hypothalamic leptin resistance and inflammation were also attenuated, allowing better appetite control in mice fed a HFD and leading to reduced food intake. Additionally, metabolic flexibility in the eWAT was improved, and the gut microbiome was modulated by low-dose valine supplementation. CONCLUSION Low-dose valine supplementation attenuates MASLD by enhancing systemic leptin sensitivity and modulating the gut microbiome.
Collapse
Affiliation(s)
- Felicianna
- School of Biological Sciences, University of Hong Kong, Pokfulam 999077, Hong Kong, China
| | - Emily K K Lo
- School of Biological Sciences, University of Hong Kong, Pokfulam 999077, Hong Kong, China
| | - Congjia Chen
- School of Biological Sciences, University of Hong Kong, Pokfulam 999077, Hong Kong, China
| | - Marsena J Ismaiah
- School of Biological Sciences, University of Hong Kong, Pokfulam 999077, Hong Kong, China
| | - Fangfei Zhang
- School of Biological Sciences, University of Hong Kong, Pokfulam 999077, Hong Kong, China
| | - Hoi Kit Matthew Leung
- School of Biological Sciences, University of Hong Kong, Pokfulam 999077, Hong Kong, China
| | - Hani El-Nezami
- School of Biological Sciences, University of Hong Kong, Pokfulam 999077, Hong Kong, China; Institute of Public Health and Clinical Nutrition, School of Medicine, University of Eastern Finland, FI-70211 Kuopio, Finland.
| |
Collapse
|
31
|
Du L, Ding X, Tian Y, Chen J, Li W. Effect of anthocyanins on metabolic syndrome through interacting with gut microbiota. Pharmacol Res 2024; 210:107511. [PMID: 39577753 DOI: 10.1016/j.phrs.2024.107511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 10/22/2024] [Accepted: 11/16/2024] [Indexed: 11/24/2024]
Abstract
Metabolic syndrome, as a complex pathological condition, is caused by a series of pathogenic factors and has become a global public health challenge. Anthocyanins, a natural water-soluble flavonoid pigment, have attracted much attention due to their antioxidant, anti-inflammatory, and anticancer biological activities. After ingestion, a majority of anthocyanins is not directly absorbed but rather reaches the colon. Hence, the exertion of their biological benefits is closely intertwined with the role played by gut microbiota. In this review, we introduce the pathogenesis and intervention methods of metabolic syndrome, as well as the interaction between anthocyanins and gut microbiota. We also discuss the therapeutic potential of anthocyanins through gut microbiota in addressing a range of metabolic syndrome conditions, including obesity, type 2 diabetes mellitus, cardiovascular diseases, non-alcoholic fatty liver disease, inflammatory bowel disease, polycystic ovary syndrome, osteoporosis, and cancer.
Collapse
Affiliation(s)
- Lanlan Du
- State Key Laboratory of Tree Genetics and Breeding, Co-Innovation Center for Sustainable Forestry in Southern China, College of Forestry and Grassland, Nanjing Forestry University, Nanjing 210037, China
| | - Xiaoqin Ding
- Jiangsu Key Laboratory for the Research and Utilization of Plant Resources, Institute of Botany, Jiangsu Province and Chinese Academy of Sciences, Nanjing 210014, China
| | - Yuwen Tian
- Jiangsu Key Laboratory for the Research and Utilization of Plant Resources, Institute of Botany, Jiangsu Province and Chinese Academy of Sciences, Nanjing 210014, China
| | - Jian Chen
- Jiangsu Key Laboratory for the Research and Utilization of Plant Resources, Institute of Botany, Jiangsu Province and Chinese Academy of Sciences, Nanjing 210014, China.
| | - Weilin Li
- State Key Laboratory of Tree Genetics and Breeding, Co-Innovation Center for Sustainable Forestry in Southern China, College of Forestry and Grassland, Nanjing Forestry University, Nanjing 210037, China.
| |
Collapse
|
32
|
Zhang H, Liu S, Ma Z, Huang H, Zheng L, Tian Y, Zhong Q. Effectively Enhancing the Physiological Activity and Sensory Quality of Whole Calamondin Puree via Yeast Fermentation. Int J Mol Sci 2024; 25:11984. [PMID: 39596054 PMCID: PMC11593417 DOI: 10.3390/ijms252211984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 10/31/2024] [Accepted: 11/04/2024] [Indexed: 11/28/2024] Open
Abstract
To explore the feasibility of pure yeast fermentation in whole Calamondin puree (FWCP) for the utilization of the whole fruit, yeasts were isolated from naturally fermented Calamondin, and their fermentation characteristics were evaluated. The results indicated that all yeasts were able to ferment FWCP, reducing the sour taste by degrading citric acid, increasing the contents of nutrients (such as phenols and limonins) and volatile compounds, and enhancing the antioxidant activity and inhibition of α-glucosidase activity (p < 0.05). Among them, P. terricola QJJY1 and H. opuntiae QJJY14 exhibited stronger abilities to degrade organic acids, with P. terricola QJJY1 enhancing the antioxidant capacity by releasing phenolic compounds such as phloretin-3',5'-di-c-β-glucoside, vitexin-2″-O-rhamnoside, and isomargaritene. Moreover, H. opuntiae QJJY15 improved the contents of characteristic volatile compounds such as terpene hydrocarbons and higher alcohols. In total, 70 components were identified as differential metabolites based on their fold change in the metabolites, with 42 differential metabolites involved in 29 metabolic pathways across four strains. The main pathways related to phenol and flavor enrichment were flavonoid, flavone, and flavonol biosynthesis, monoterpenoid biosynthesis, and glyoxylate and dicarboxylate metabolism. Therefore, yeast fermentation is an effective method for utilizing whole Calamondin.
Collapse
Affiliation(s)
- Hongjian Zhang
- Hainan University-HSF/LWL Collaborative Innovation Laboratory, College of Food Sciences & Engineering, Hainan University, Haikou 570228, China
- Hainan Institute of Grain and Oil Science, Qionghai 571400, China
| | - Shuaiguang Liu
- Hainan Institute of Grain and Oil Science, Qionghai 571400, China
| | - Zewei Ma
- Hainan Institute of Grain and Oil Science, Qionghai 571400, China
| | - Huan Huang
- Hainan Institute of Grain and Oil Science, Qionghai 571400, China
| | - Lianhe Zheng
- Hainan Institute of Grain and Oil Science, Qionghai 571400, China
| | - Yan Tian
- Engineering Research Center of Utilization of Tropical Polysaccharide Resources (Ministry of Education), College of Food Science and Engineering, Hainan University, Haikou 570228, China
| | - Qiuping Zhong
- Hainan University-HSF/LWL Collaborative Innovation Laboratory, College of Food Sciences & Engineering, Hainan University, Haikou 570228, China
| |
Collapse
|
33
|
Li Z, Chen L, Qu L, Yu W, Liu T, Ning F, Li J, Guo X, Sun F, Sun B, Luo L. Potential implications of natural compounds on aging and metabolic regulation. Ageing Res Rev 2024; 101:102475. [PMID: 39222665 DOI: 10.1016/j.arr.2024.102475] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 08/12/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
Aging is generally accompanied by a progressive loss of metabolic homeostasis. Targeting metabolic processes is an attractive strategy for healthy-aging. Numerous natural compounds have demonstrated strong anti-aging effects. This review summarizes recent findings on metabolic pathways involved in aging and explores the anti-aging effects of natural compounds by modulating these pathways. The potential anti-aging effects of natural extracts rich in biologically active compounds are also discussed. Regulating the metabolism of carbohydrates, proteins, lipids, and nicotinamide adenine dinucleotide is an important strategy for delaying aging. Furthermore, phenolic compounds, terpenoids, alkaloids, and nucleotide compounds have shown particularly promising effects on aging, especially with respect to metabolism regulation. Moreover, metabolomics is a valuable tool for uncovering potential targets against aging. Future research should focus on identifying novel natural compounds that regulate human metabolism and should delve deeper into the mechanisms of metabolic regulation using metabolomics methods, aiming to delay aging and extend lifespan.
Collapse
Affiliation(s)
- Zhuozhen Li
- Key Laboratory of Geriatric Nutrition and Health of Ministry of Education, School of Food and Health, Beijing Technology and Business University, Beijing 100048, China
| | - Lili Chen
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China; School of Life Science, Jiangxi Science & Technology Normal University, Nanchang 330013, China
| | - Liangliang Qu
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China
| | - Wenjie Yu
- Key Laboratory of Geriatric Nutrition and Health of Ministry of Education, School of Food and Health, Beijing Technology and Business University, Beijing 100048, China
| | - Tao Liu
- Key Laboratory of Geriatric Nutrition and Health of Ministry of Education, School of Food and Health, Beijing Technology and Business University, Beijing 100048, China
| | - Fangjian Ning
- Key Laboratory of Geriatric Nutrition and Health of Ministry of Education, School of Food and Health, Beijing Technology and Business University, Beijing 100048, China
| | - Jinwang Li
- Key Laboratory of Geriatric Nutrition and Health of Ministry of Education, School of Food and Health, Beijing Technology and Business University, Beijing 100048, China
| | - Xiali Guo
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China
| | - Fengjie Sun
- Department of Biological Sciences, School of Science and Technology, Georgia Gwinnett College, Lawrenceville, GA 30043, USA
| | - Baoguo Sun
- Key Laboratory of Geriatric Nutrition and Health of Ministry of Education, School of Food and Health, Beijing Technology and Business University, Beijing 100048, China
| | - Liping Luo
- Key Laboratory of Geriatric Nutrition and Health of Ministry of Education, School of Food and Health, Beijing Technology and Business University, Beijing 100048, China; State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China.
| |
Collapse
|
34
|
Liu Y, Yan N, Chen Q, Dong L, Li Y, Weng P, Wu Z, Pan D, Liu L, Farag MA, Wang L, Liu L. Research advances in citrus polyphenols: green extraction technologies, gut homeostasis regulation, and nano-targeted delivery system application. Crit Rev Food Sci Nutr 2024; 64:11493-11509. [PMID: 37552798 DOI: 10.1080/10408398.2023.2239350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/10/2023]
Abstract
Citrus polyphenols can modulate gut microbiota and such bi-directional interaction that can yield metabolites such as short-chain fatty acids (SCFAs) to aid in gut homeostasis. Such interaction provides citrus polyphenols with powerful prebiotic potential, contributing to guts' health status and metabolic regulation. Citrus polyphenols encompass unique polymethoxy flavonoids imparting non-polar nature that improve their bioactivities and ability to penetrate the blood-brain barrier. Green extraction technology targeting recovery of these polyphenols has received increasing attention due to its advantages of high extraction yield, short extraction time, low solvent consumption, and environmental friendliness. However, the low bioavailability of citrus polyphenols limits their applications in extraction from citrus by-products. Meanwhile, nano-encapsulation technology may serve as a promising approach to improve citrus polyphenols' bioavailability. As citrus polyphenols encompass multiple hydroxyl groups, they are potential to interact with bio-macromolecules such as proteins and polysaccharides in nano-encapsulated systems that can improve their bioavailability. This multifaceted review provides a research basis for the green and efficient extraction techniques of citrus polyphenols, as well as integrated mechanisms for its anti-inflammation, alleviating metabolic syndrome, and regulating gut homeostasis, which is more capitalized upon using nano-delivery systems as discussed in that review to maximize their health and food applications.
Collapse
Affiliation(s)
- Yahui Liu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Animal Protein Deep Processing Technology of Zhejiang, Zhejiang-Malaysia Joint Research Laboratory for Agricultural Product Processing and Nutrition, School of Food and Pharmaceutical Sciences, Ningbo University, Ningbo, Zhejiang, China
| | - Ning Yan
- Plant Functional Component Research Center, Tobacco Research Institute of Chinese Academy of Agricultural Sciences, Laoshan District, Qingdao, China
| | - Qin Chen
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Animal Protein Deep Processing Technology of Zhejiang, Zhejiang-Malaysia Joint Research Laboratory for Agricultural Product Processing and Nutrition, School of Food and Pharmaceutical Sciences, Ningbo University, Ningbo, Zhejiang, China
| | - Lezhen Dong
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Animal Protein Deep Processing Technology of Zhejiang, Zhejiang-Malaysia Joint Research Laboratory for Agricultural Product Processing and Nutrition, School of Food and Pharmaceutical Sciences, Ningbo University, Ningbo, Zhejiang, China
| | - Ying Li
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Animal Protein Deep Processing Technology of Zhejiang, Zhejiang-Malaysia Joint Research Laboratory for Agricultural Product Processing and Nutrition, School of Food and Pharmaceutical Sciences, Ningbo University, Ningbo, Zhejiang, China
| | - Peifang Weng
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Animal Protein Deep Processing Technology of Zhejiang, Zhejiang-Malaysia Joint Research Laboratory for Agricultural Product Processing and Nutrition, School of Food and Pharmaceutical Sciences, Ningbo University, Ningbo, Zhejiang, China
| | - Zufang Wu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Animal Protein Deep Processing Technology of Zhejiang, Zhejiang-Malaysia Joint Research Laboratory for Agricultural Product Processing and Nutrition, School of Food and Pharmaceutical Sciences, Ningbo University, Ningbo, Zhejiang, China
| | - Daodong Pan
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Animal Protein Deep Processing Technology of Zhejiang, Zhejiang-Malaysia Joint Research Laboratory for Agricultural Product Processing and Nutrition, School of Food and Pharmaceutical Sciences, Ningbo University, Ningbo, Zhejiang, China
| | - Lingyi Liu
- Department of Food Science and Technology, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
| | - Mohamed A Farag
- Pharmacognosy Department, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Lei Wang
- School of Liquor and Food Engineering, Guizhou University, Guiyang, Guizhou, China
| | - Lianliang Liu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Animal Protein Deep Processing Technology of Zhejiang, Zhejiang-Malaysia Joint Research Laboratory for Agricultural Product Processing and Nutrition, School of Food and Pharmaceutical Sciences, Ningbo University, Ningbo, Zhejiang, China
| |
Collapse
|
35
|
MORALES-CANO KL, RIVERA-ALAMILLO YC, OLIART-ROS RM, PEÑA-MONTES C. Modulation of the gut microbiota by dietary intervention with Acanthocereus tetragonus improves the health status of Wistar rats with metabolic syndrome. BIOSCIENCE OF MICROBIOTA, FOOD AND HEALTH 2024; 44:100-109. [PMID: 40171391 PMCID: PMC11957756 DOI: 10.12938/bmfh.2024-041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 09/15/2024] [Indexed: 04/03/2025]
Abstract
The gastrointestinal tract is an ecosystem with heterogeneous patterns, distributions, and environments, resulting in different microbial compositions in each gut segment. The relationship between diet and microbiota determines this heterogeneity. Consumption of diets high in fat and carbohydrates (HLHC) is associated with gut dysbiosis, low microbial diversity, and metabolic syndrome (MetS). Functional fiber consumption improves the profile and diversity of the gut microbiota (GM); it stimulates the production of short-chain fatty acids (SCFAs), which act as signaling molecules that maintain the gut barrier integrity and induce hormone synthesis that regulates satiety and glucose metabolism, reducing some MetS parameters. The effect of a dietary intervention with Acanthocereus tetragonus (At), a cactus rich in fiber, antioxidants, amino acids, and minerals traditionally consumed by the Mexican population, is reported here. For this purpose, Wistar rats were randomly divided into three study groups: a control (C) group, a MetS group, and an At-supplemented group. In the MetS and At groups, an HLHC was administered for 12 weeks, inducing MetS. After 18 weeks, stool samples were collected for microbiota sequencing. HLHC administration favored Firmicutes and decreased the abundance of Bacteriodetes at the phylum level in the MetS group. At the genus level, the dietary intervention with At increased the presence of Roseburia, Ruminococcus, Blautia, Bacteroides, and Christensenella, reflecting the effect of A. tetragonus consumption on GM. At diet administration reduced body weight; the plasma glucose, insulin, and lipid levels; and insulin resistance.
Collapse
Affiliation(s)
- Karla Lizzeth MORALES-CANO
- National Technological Institute of Mexico Campus Veracruz,
Miguel Ángel de Quevedo 2779, Formando Hogar, Veracruz, Ver. CP 91897, Mexico
| | - Yokebed Cecilia RIVERA-ALAMILLO
- National Technological Institute of Mexico Campus Veracruz,
Miguel Ángel de Quevedo 2779, Formando Hogar, Veracruz, Ver. CP 91897, Mexico
| | - Rosa Maria OLIART-ROS
- National Technological Institute of Mexico Campus Veracruz,
Miguel Ángel de Quevedo 2779, Formando Hogar, Veracruz, Ver. CP 91897, Mexico
| | - Carolina PEÑA-MONTES
- National Technological Institute of Mexico Campus Veracruz,
Miguel Ángel de Quevedo 2779, Formando Hogar, Veracruz, Ver. CP 91897, Mexico
| |
Collapse
|
36
|
Si ZL, Wang HY, Wang T, Cao YZ, Li QZ, Liu K, Huang Z, Liu HL, Tan YJ, Wang YY, Huang FQ, Ma GX, Alolga RN, Yan M, Chen C, Li JH, Li J, Liu HW, Zhang ZH. Gut Bacteroides ovatus ameliorates renal fibrosis by promoting the production of HDCA through upregulation of Clostridium scindens. Cell Rep 2024; 43:114830. [PMID: 39392759 DOI: 10.1016/j.celrep.2024.114830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 07/14/2024] [Accepted: 09/19/2024] [Indexed: 10/13/2024] Open
Abstract
Renal fibrosis, inflammation, and gut dysbiosis are all linked to chronic kidney disease (CKD). Here we show that Bacteroides ovatus protects against renal fibrosis. Mechanistically, B. ovatus enhances intestinal hyodeoxycholic acid (HDCA) levels by upregulating a strain of intestinal bacteria, Clostridium scindens, that has the capacity for direct HDCA production in mice. HDCA significantly promoted GLP-1 secretion by upregulating the expression of TGR5 and downregulating the expression of farnesoid X receptor (FXR) in the gut. Activation of renal GLP-1R attenuates renal fibrosis while delaying the subsequent development of CKD. In addition, HDCA can also protect against renal fibrosis by directly upregulating renal TGR5. The natural product neohesperidin (NHP) was found to exert its anti-renal fibrotic effects by promoting the growth of B. ovatus. Our findings provide mechanistic insights into the therapeutic potential of B. ovatus, C. scindens, and HDCA in treating CKD.
Collapse
Affiliation(s)
- Zi-Lin Si
- Key Laboratory of Tropical Biological Resources of the Ministry of Education and One Health Institute, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China; School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Han-Yu Wang
- Key Laboratory of Tropical Biological Resources of the Ministry of Education and One Health Institute, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China; State Key Laboratory of Natural Medicines, Department of TCM Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Tao Wang
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, No. 1 Beichenxi Road, Chaoyang District, Beijing 100101, P.R. China
| | - Yi-Zhi Cao
- Key Laboratory of Tropical Biological Resources of the Ministry of Education and One Health Institute, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China; State Key Laboratory of Natural Medicines, Department of TCM Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Qing-Zhen Li
- Key Laboratory of Tropical Biological Resources of the Ministry of Education and One Health Institute, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China; State Key Laboratory of Natural Medicines, Department of TCM Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Kang Liu
- Department of Nephrology, Jiangsu Province Hospital (The First Affiliated Hospital of Nanjing Medical University), Nanjing 210029, China
| | - Zhou Huang
- Key Laboratory of Tropical Biological Resources of the Ministry of Education and One Health Institute, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China; State Key Laboratory of Natural Medicines, Department of TCM Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Hui-Ling Liu
- Key Laboratory of Tropical Biological Resources of the Ministry of Education and One Health Institute, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China
| | - Ya-Jie Tan
- State Key Laboratory of Natural Medicines, Department of TCM Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Yin-Yin Wang
- State Key Laboratory of Natural Medicines, Department of TCM Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Feng-Qing Huang
- State Key Laboratory of Natural Medicines, Department of TCM Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Gao-Xiang Ma
- State Key Laboratory of Natural Medicines, Department of TCM Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Raphael N Alolga
- State Key Laboratory of Natural Medicines, Department of TCM Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Miao Yan
- Department of Nephrology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Cheng Chen
- Department of Nephrology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Jun-Hui Li
- Putuo People's Hospital, Tongji University, Shanghai 200060, China
| | - Jing Li
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Hong-Wei Liu
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, No. 1 Beichenxi Road, Chaoyang District, Beijing 100101, P.R. China
| | - Zhi-Hao Zhang
- Key Laboratory of Tropical Biological Resources of the Ministry of Education and One Health Institute, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China; State Key Laboratory of Natural Medicines, Department of TCM Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China.
| |
Collapse
|
37
|
Qian X, Lin X, Hu W, Zhang L, Chen W, Zhang S, Ge S, Xu X, Luo K. Intestinal homeostasis disrupted by Periodontitis exacerbates Alzheimer's Disease in APP/PS1 mice. J Neuroinflammation 2024; 21:263. [PMID: 39425119 PMCID: PMC11489998 DOI: 10.1186/s12974-024-03256-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 10/07/2024] [Indexed: 10/21/2024] Open
Abstract
Periodontitis exacerbates Alzheimer's disease (AD) through multiple pathways. Both periodontitis and AD are intricately correlated to intestinal homeostasis, yet there is still a lack of direct evidence regarding whether periodontitis can regulate the progression of AD by modulating intestinal homeostasis. The current study induced experimental periodontitis in AD mice by bilaterally ligating the maxillary second molars with silk and administering Pg-LPS injections in APPswe/PS1ΔE9 (APP/PS1) mice. Behavioral tests and histological analyses of brain tissue were conducted after 8 weeks. Gut microbiota was analyzed and colon tissue were also evaluated. Then, fecal microbiota from mice with periodontitis was transplanted into antibiotic-treated mice to confirm the effects of periodontitis on AD and the potential mechanism was explored. The results indicated periodontitis exacerbated cognitive impairment and anxious behaviour in APP/PS1 mice, with increased Aβ deposition, microglial overactivation and neuroinflammation in brain. Moreover, the intestinal homeostasis of AD mice was altered by periodontitis, including affecting gut microbiota composition, causing colon inflammation and destroyed intestinal epithelial barrier. Furthermore, AD mice that underwent fecal transplantation from mice with periodontitis exhibited worsened AD progression and disrupted intestinal homeostasis. It also impaired intestinal barrier function, elevated peripheral inflammation, damaged blood-brain barrier (BBB) and caused neuroinflammation and synapses impairment. Taken together, the current study demonstrated that periodontitis could disrupt intestinal homeostasis to exacerbate AD progression potential via causing gut microbial dysbiosis, intestinal inflammation and intestinal barrier impairment to induce peripheral inflammation and damage BBB, ultimately leading to neuroinflammation and synapse impairment. It underscores the importance of maintaining both periodontal health and intestinal homeostasis to reduce the risk of AD.
Collapse
Affiliation(s)
- Xueshen Qian
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, P.R. China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350002, P.R. China
| | - Xuxin Lin
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, P.R. China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350002, P.R. China
| | - Weiqiang Hu
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, P.R. China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350002, P.R. China
| | - Lu Zhang
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, P.R. China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350002, P.R. China
| | - Wenqian Chen
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, P.R. China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350002, P.R. China
| | - Shuang Zhang
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Institute of Stomatology, Nanjing University, Nanjing, Jiangsu, 210008, P.R. China
| | - Song Ge
- School and Hospital of Stomatology, Zunyi Medical University, Zunyi, Guizhou, 563003, P.R. China
| | - Xiongcheng Xu
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, P.R. China.
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350002, P.R. China.
| | - Kai Luo
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, P.R. China.
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350002, P.R. China.
| |
Collapse
|
38
|
Mo X, Cheng R, Shen L, Liu N, Sun Y, Lin S, Jiang G, Li X, Peng X, Zhang Y, Liao Y, Yan H, Liu L. Yeast β-glucan alleviates high-fat diet-induced Alzheimer's disease-like pathologies in rats via the gut-brain axis. Int J Biol Macromol 2024; 278:134939. [PMID: 39179066 DOI: 10.1016/j.ijbiomac.2024.134939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 05/15/2024] [Accepted: 08/20/2024] [Indexed: 08/26/2024]
Abstract
Targeting the gut microbiota may be an emerging strategy for the prevention and treatment of Alzheimer's disease (AD). Macro-molecular yeast β-glucan (BG), derived from the yeast of Saccharomyces cerevisiae, regulates the gut microbiota. This study aimed to investigate the effect and mechanism of long-term BG in high-fat diet (HFD)-induced AD-like pathologies from the perspective of the gut microbiota. Here, we found that 80 weeks of BG treatment ameliorated HFD-induced cognitive dysfunction in rats. In the hippocampus, BG alleviated HFD-induced the activation of astrocytes, microglia, NOD-like receptor thermal protein domain associated protein 3 (NLRP3) inflammasome pathway, and AD-like pathologies. BG modulated gut dysbiosis through increasing the levels of beneficial bacteria and short-chain fatty acids (SCFAs). BG also attenuated HFD-induced gut barrier impairment. Correlation analysis revealed a close relationship among microbiota, SCFAs, and AD-like pathologies. Furthermore, the fecal microbiota of BG-treated rats and SCFAs treatment mitigated AD-like pathologies via the NLRP3 inflammasome pathway in HFD-fed aged rats. These results suggested that long-term BG promotes the production of SCFAs derived from gut microbiota, which further inhibits NLRP3 inflammasome-mediated neuroinflammation, thereby alleviating HFD-induced AD-like pathologies in rats. BG may become a new strategy for targeting neurodegenerative diseases.
Collapse
Affiliation(s)
- Xiaoxing Mo
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan 430030, China.
| | - Ruijie Cheng
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan 430030, China.
| | - Lihui Shen
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan 430030, China.
| | - Nian Liu
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan 430030, China
| | - Yunhong Sun
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan 430030, China.
| | - Shan Lin
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan 430030, China.
| | - Guanhua Jiang
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan 430030, China.
| | - Xiaoqin Li
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan 430030, China.
| | - Xiaobo Peng
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan 430030, China.
| | - Yan Zhang
- The Hubei Provincial Key Laboratory of Yeast Function, Yichang 443003, China.
| | - Yuxiao Liao
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan 430030, China.
| | - Hong Yan
- Department of Health Toxicology, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan 430030, China.
| | - Liegang Liu
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan 430030, China.
| |
Collapse
|
39
|
Zhou SK, Xu JD, Gao XQ, Zhang RJ, Cheng FF, Yao WF, Zhang Y, Geng T, Zhang L. Fructus Jujubae cooperated with water-expelling members in Shizao decoction alleviated intestinal injury and malignant ascites by modulating gut microbiota and metabolic homeostasis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 133:155895. [PMID: 39084184 DOI: 10.1016/j.phymed.2024.155895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 05/11/2024] [Accepted: 07/15/2024] [Indexed: 08/02/2024]
Abstract
BACKGROUND Shizao decoction (SZD) consisted of Euphorbia kansui (EK), Euphorbia pekinensis (EP), Daphne genkwa (DG), and Fructus Jujubae (FJ) is a classic Chinese herbal medicine formula for treating malignant ascites, which is closely related to the modulation of gut microbiota by our previous study. For water-expelling members (WEM) including EK, EP, and DG may have side effects on the intestine, FJ is employed for detoxification and effectivity enhancement of WEM. However, the underlying mechanism for the compatibility of WEM and FJ is still unknown. PURPOSE To investigate the effect of the compatibility of WEM with FJ in SZD on malignant ascites and elucidate the potential mechanism from the perspective of the modulation of gut microbiota and related metabolic function. METHODS Qualitative and quantitative evaluation of main components was conducted for comprehensive characterization of SZD and WEM. The effect of WEM and SZD was compared on malignant ascites effusion (MAE) rats. The intestinal injury was evaluated by HE staining and oxidative damage. Ascites weight, urine amount, fecal water content, the expression of aquaporins, and cytokines in ascites (IL-6, VEGF, and TNF-α) were measured to estimate the water-expelling activity. The intestinal flora was detected by 16S rDNA sequencing and the content of fecal short-chain fatty acids (SCFAs) was analyzed using gas chromatography-mass spectrometry. Pseudo-germ-free (PGF) and fecal bacteria transplantation animal experiments were subsequently employed to validate this finding. The fecal metabolomics and correlation analysis were finally conducted to explore the related metabolic changes. RESULTS 51 and 33 components were identified in SZD and WEM, respectively. Compared to WEM alone, the compatibility with FJ remarkably reduced intestinal oxidative damage in MAE rats. Ascites was also relieved by downregulating the expression of AQP3 in the colon and decreasing the levels of IL-6, TNF-α and VEGF in ascites. The diversity of gut microbiota was reversed with an increase in Lactobacillus and Clostridia_UCG-014 while a decrease in Colidextribacter. Under the PGF condition, compatibility of WEM with FJ failed to reduce intestinal injury and alleviate MA significantly, but this effect was further enhanced after FMT. 23 potential fecal metabolites were finally identified. Correlation analysis further showed that Lactobacillus and Clostridia_UCG-014 were positively correlated with SCFAs and l-tryptophan. Colidextribacter was negatively correlated with thymidine but positively correlated with ursodeoxycholic acid and deoxycholic acid. CONCLUSION FJ cooperated with WEM reduced intestinal injury and alleviated malignant ascites by modulating gut microbiota, short-chain fatty and tryptophan metabolism. These findings provide a scientific basis for the clinical application of FJ from SZD and the safe usage of SZD.
Collapse
Affiliation(s)
- Shi-Kang Zhou
- Jiangsu Key Laboratory for High Technology Research of TCM Formulae, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine and Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, No.138, Xianlin Road, Qixia District, Nanjing 210023, PR China; Changzhou Key Laboratory of Human Use Experience Research & Transformation of Menghe Medical Sect, Changzhou Hospital of Chinese Medicinal Affiliated to Nanjing University of Chinese Medicine, No. 25 Heping North Road, Tianning District, Changzhou, 213003, PR China
| | - Jin-Di Xu
- Jiangsu Key Laboratory for High Technology Research of TCM Formulae, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine and Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, No.138, Xianlin Road, Qixia District, Nanjing 210023, PR China
| | - Xiao-Qin Gao
- Jiangsu Key Laboratory for High Technology Research of TCM Formulae, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine and Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, No.138, Xianlin Road, Qixia District, Nanjing 210023, PR China
| | - Ren-Jie Zhang
- Jiangsu Key Laboratory for High Technology Research of TCM Formulae, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine and Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, No.138, Xianlin Road, Qixia District, Nanjing 210023, PR China
| | - Fang-Fang Cheng
- Jiangsu Key Laboratory for High Technology Research of TCM Formulae, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine and Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, No.138, Xianlin Road, Qixia District, Nanjing 210023, PR China
| | - Wei-Feng Yao
- Jiangsu Key Laboratory for High Technology Research of TCM Formulae, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine and Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, No.138, Xianlin Road, Qixia District, Nanjing 210023, PR China; Taizhou Key Laboratory for Development of Traditional Chinese Medicine Health Products, Taizhou Engineering Research Center for Quality and Industrialization of Traditional Chinese Medicine, Nanjing University of Chinese Medicine Hanlin College, No.6, Kuangshi Road, Pharmaceutical High-tech District, Taizhou 225300, PR China
| | - Yi Zhang
- Jiangsu Key Laboratory for High Technology Research of TCM Formulae, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine and Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, No.138, Xianlin Road, Qixia District, Nanjing 210023, PR China.
| | - Ting Geng
- Jiangsu Key Laboratory for High Technology Research of TCM Formulae, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine and Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, No.138, Xianlin Road, Qixia District, Nanjing 210023, PR China; Taizhou Key Laboratory for Development of Traditional Chinese Medicine Health Products, Taizhou Engineering Research Center for Quality and Industrialization of Traditional Chinese Medicine, Nanjing University of Chinese Medicine Hanlin College, No.6, Kuangshi Road, Pharmaceutical High-tech District, Taizhou 225300, PR China.
| | - Li Zhang
- Jiangsu Key Laboratory for High Technology Research of TCM Formulae, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine and Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, No.138, Xianlin Road, Qixia District, Nanjing 210023, PR China; Taizhou Key Laboratory for Development of Traditional Chinese Medicine Health Products, Taizhou Engineering Research Center for Quality and Industrialization of Traditional Chinese Medicine, Nanjing University of Chinese Medicine Hanlin College, No.6, Kuangshi Road, Pharmaceutical High-tech District, Taizhou 225300, PR China.
| |
Collapse
|
40
|
Wang J, Zhang Z, Dai T, Zhang Z, Zhang Q, Yao J, Wang L, He N, Li S. The therapeutic effect and possible mechanisms of alginate oligosaccharide on metabolic syndrome by regulating gut microbiota. Food Funct 2024; 15:9632-9661. [PMID: 39239698 DOI: 10.1039/d4fo02802c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/07/2024]
Abstract
Metabolic syndrome (MetS) is a disease condition incorporating the abnormal accumulation of various metabolic components, including overweight or abdominal obesity, insulin resistance and abnormal glucose tolerance, hypertension, atherosclerosis, or dyslipidemia. It has been proved that the gut microbiota and microbial-derived products play an important role in regulating lipid metabolism and thus the onset and development of MetS. Previous studies have demonstrated that oligosaccharides with prebiotic effects, such as chitosan oligosaccharides, can regulate the structure of the microbial community and its derived products to control weight and reduce MetS associated with obesity. Alginate oligosaccharides (AOS), natural products extracted from degraded alginate salts with high solubility and extensive biological activity, have also been found to modulate gut microbiota. This review aims to summarize experimental evidence on the positive effects of AOS on different types of MetS while providing insights into mechanisms through which AOS regulates gut microbiota for preventing and treating MetS.
Collapse
Affiliation(s)
- Jingyi Wang
- School of Basic Medicine, Qingdao Medical College, Qingdao, University, Qingdao 266071, China.
- Department of Obstetrics and Gynecology, The Affiliated Qingdao Municipal Hospital of Qingdao University, Qingdao, 266000, China
| | - Zixuan Zhang
- School of Basic Medicine, Qingdao Medical College, Qingdao, University, Qingdao 266071, China.
| | - Tong Dai
- School of Basic Medicine, Qingdao Medical College, Qingdao, University, Qingdao 266071, China.
| | - Ziheng Zhang
- School of Basic Medicine, Qingdao Medical College, Qingdao, University, Qingdao 266071, China.
| | - Qingfeng Zhang
- School of Basic Medicine, Qingdao Medical College, Qingdao, University, Qingdao 266071, China.
| | - Jingtong Yao
- School of Basic Medicine, Qingdao Medical College, Qingdao, University, Qingdao 266071, China.
| | - Lijing Wang
- School of Basic Medicine, Qingdao Medical College, Qingdao, University, Qingdao 266071, China.
- Department of Obstetrics and Gynecology, The Affiliated Qingdao Municipal Hospital of Qingdao University, Qingdao, 266000, China
- Department of Obstetrics, Qingdao Municipal Hospital, Qingdao, 266000, China
| | - Ningning He
- School of Basic Medicine, Qingdao Medical College, Qingdao, University, Qingdao 266071, China.
| | - Shangyong Li
- School of Basic Medicine, Qingdao Medical College, Qingdao, University, Qingdao 266071, China.
| |
Collapse
|
41
|
Zhu LY, Zhang MY, Juan-Cheng, Zhang YX. Shield-armed probiotic delivery system based on co-deposition of poly-dopamine and poly-lysine helps Lactiplantibacillus plantarum relieve hyperuricemia. Int J Biol Macromol 2024; 280:135666. [PMID: 39299415 DOI: 10.1016/j.ijbiomac.2024.135666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 08/28/2024] [Accepted: 09/12/2024] [Indexed: 09/22/2024]
Abstract
Hyperuricemia (HUA) is a disease characterized by an abnormal metabolism of purine. Lactic acid bacteria (LAB) have attracted much attention for their safe and effective treatment of HUA by inhibiting xanthine oxidase (XOD) and regulating gut microbiota. However, the effectiveness of probiotics can be compromised by the harsh environment of the gastrointestinal tract. In preliminary experiments, Lactiplantibacillus plantarum DY1, which is generally regarded as safe (GRAS), can lower uric acid. We have devised a straightforward and efficient technique for encapsulating DY1 using a coating comprising polydopamine (PDA) co-deposited with poly-l-lysine (PLL) to obtain DY1@PDLL. TEM, SEM, FT-IR and DLS tests showed that DY1 was successfully coated. Incubate at SGF or SIF for 3 h, the number of viable bacteria of free probiotics and DY1@PDLL decreased by 0.92 and 0.46 log cfu/mL, 1.66 and 0.66 log cfu/mL, respectively. The fluorescence intensity of the intestines of the DY1@PDLL treated mice was 3.96 times that of free probiotic. Notably, DY1@PDLL can reduce the uric acid levels of HUA mice by 31.63 % and free probiotics by 18.72 % (≈1.69 times). DY1@PDLL could also regulate gut microbiota and serum metabolic profile. These findings unequivocally highlight the remarkable potential of DY1@PDLL as an exceptional oral probiotic delivery system.
Collapse
Affiliation(s)
- Lin-Yan Zhu
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Meng-Yue Zhang
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Juan-Cheng
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Yi-Xuan Zhang
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China.
| |
Collapse
|
42
|
Dong B, Peng Y, Wang M, Peng C, Li X. Multi-omics integrated analyses indicated that non-polysaccharides of Sijunzi decoction ameliorated spleen deficiency syndrome via regulating microbiota-gut-metabolites axis and exerted synergistic compatibility. JOURNAL OF ETHNOPHARMACOLOGY 2024; 331:118276. [PMID: 38697408 DOI: 10.1016/j.jep.2024.118276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 04/26/2024] [Accepted: 04/29/2024] [Indexed: 05/05/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE As a classical traditional Chinese medicine formula to invigorating spleen and replenishing qi, Sijunzi decoction (SJZD) is composed of four herbs, which is applied to cure spleen deficiency syndrome (SDS) clinically. The non-polysaccharides (NPSs) of SJZD (SJZD_NPS) are important pharmacodynamic material basis. However, the amelioration mechanism of SJZD_NPS on SDS has not been fully elaborated. Additionally, the contribution of herbs compatibility to efficacy of this formula remains unclear. AIM OF THE STUDY The aim was to explore the underlying mechanisms of SJZD_NPS on improving SDS, and uncover the scientific connotation in SJZD compatibility. MATERIALS AND METHODS A strategy integrating incomplete formulae (called "Chai-fang" in Chinese) comparison, pharmacodynamics, gut microbiome, and metabolome was employed to reveal the role of each herb to SJZD compatibility against SDS. Additionally, the underlying mechanism harbored by SJZD_NPS was further explored through targeted metabolomics, network pharmacology, molecular docking, pseudo-sterile model, and metagenomics. RESULTS SJZD_NPS significantly alleviated diarrhea, disordered secretion of gastrointestinal hormones and neurotransmitters, damage of ileal morphology and intestinal barrier in SDS rats, which was superior to the NPSs of Chai-fang. 16S rRNA gene sequencing and metabolomics analyses revealed that SJZD_NPS effectively restored the disturbed gut microbiota community and abnormal metabolism caused by SDS, showing the most evident recovery. Moreover, SJZD_NPS recalled the levels of partial amino acids, short chain fatty acids and bile acids, which possessed strong binding affinity towards potential targets. The depletion of gut microbiota confirmed that the SDS-amelioration efficacy of SJZD_NPS is dependent on the intact gut microbiome, with the relative abundance of potential probiotics such as Lactobacillus_johnsonii and Lactobacillus_taiwanensis been enriched. CONCLUSION NPSs in SJZD can improve SDS-induced gastrointestinal-nervous system dysfunction through regulating microbiota-gut-metabolites axis, with four herbs exerting synergistic effects, which indicated the compatibility rationality of SJZD.
Collapse
Affiliation(s)
- Bangjian Dong
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Ying Peng
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Mengyue Wang
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Chongsheng Peng
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xiaobo Li
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China.
| |
Collapse
|
43
|
Jiang ZM, Wang FF, Zhao YY, Lu LF, Jiang XY, Huang TQ, Lin Y, Guo L, Weng ZB, Liu EH. Hypericum perforatum L. attenuates depression by regulating Akkermansia muciniphila, tryptophan metabolism and NFκB-NLRP2-Caspase1-IL1β pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 132:155847. [PMID: 38996505 DOI: 10.1016/j.phymed.2024.155847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 06/14/2024] [Accepted: 06/25/2024] [Indexed: 07/14/2024]
Abstract
BACKGROUND Gut microbiota dysbiosis significantly contributes to progression of depression. Hypericum perforatum L. (HPL) is traditionally used in Europe for treating depression. However, its mechanism remains largely underexplored. PURPOSE This study aims to investigate the pivotal gut microbiota species and microbial signaling metabolites associated with the antidepressant effects of HPL. METHODS Fecal microbiota transplantation was used to assess whether HPL mitigates depression through alterations in gut microbiota. Microbiota and metabolic profiling of control, chronic restraint stress (CRS)-induced depression, and HPL-treated CRS mice were examined using 16S rRNA gene sequencing and metabolomics analysis. The influence of gut microbiota on HPL's antidepressant effects was assessed by metabolite and bacterial intervention experiments. RESULTS HPL significantly alleviated depression symptoms in a manner dependent on gut microbiota and restored gut microbial composition by enriching Akkermansia muciniphila (AKK). Metabolomic analysis indicated that HPL regulated tryptophan metabolism, reducing kynurenine (KYN) levels derived from microbiota and increasing 5-hydroxytryptophan (5-HTP) levels. Notably, supplementation with KYN activated the NFκB-NLRP2-Caspase1-IL1β pathway and increased proinflammatory IL1β in the hippocampus of mice with depression. Interestingly, mono-colonization with AKK notably increased 5-hydroxytryptamine (5-HT) and decreased KYN levels, ameliorating depression symptoms through modulation of the NFκB-NLRP2-Caspase1-IL1β pathway. CONCLUSIONS The promising therapeutic role of HPL in treating depression is primarily attributed to its regulation of the NFκB-NLRP2-Caspase1-IL1β pathway, specifically by targeting AKK and tryptophan metabolites.
Collapse
Affiliation(s)
- Zheng-Meng Jiang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Fang-Fang Wang
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450052, China
| | - Yuan-Yuan Zhao
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Lin-Feng Lu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Xiao-Yu Jiang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Tian-Qing Huang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Yang Lin
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Long Guo
- School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, 050200 China.
| | - Ze-Bin Weng
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - E-Hu Liu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| |
Collapse
|
44
|
Aslan MN, Sukan-Karaçağıl B, Acar-Tek N. Roles of citrus fruits on energy expenditure, body weight management, and metabolic biomarkers: a comprehensive review. Nutr Rev 2024; 82:1292-1307. [PMID: 37702528 PMCID: PMC11317776 DOI: 10.1093/nutrit/nuad116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/14/2023] Open
Abstract
Citrus fruits are widely consumed for their nutritional and health benefits. They belong to the Rutaceae and have many varieties, such as sweet orange (Citrus sinensis), which is the most popular. Citrus fruits are rich in water (>80%), dietary fiber, and vitamins. They also contain bioactive components, which may modulate energy metabolism and lipid oxidation through various mechanisms. These mechanisms include stimulating β3-adrenergic receptors, increasing mitochondrial biogenesis and thermogenesis, activating AMP kinase and peroxisome proliferator-activated receptor-gamma coactivator-1α pathways, inhibiting lipogenesis and lipid accumulation, and inducing browning of white adipose tissue. This review summarizes the mechanisms and outcomes of citrus fruits and their metabolites on energy metabolism and body weight in different experimental models. The literature was searched for in vitro and in vivo animal and human studies that investigated the effects of citrus consumption on energy expenditure, thermogenesis, adipogenesis, and lipid accumulation. Citrus fruits and their metabolites have shown promising effects on energy metabolism and lipid oxidation in in vitro and in vivo animal studies. However, the evidence from human studies is limited and inconsistent. Possible reasons for the discrepancy are briefly discussed, and knowledge gaps and research needs are identified for future studies. Citrus fruits may have beneficial effects on energy metabolism and body weight, but more rigorous and well-designed human trials are needed to confirm their efficacy and safety.
Collapse
Affiliation(s)
- Merve Nur Aslan
- Faculty of Health Sciences, Department of Nutrition and Dietetics, Bolu Abant Izzet Baysal University, Bolu, Turkey
- Department of Nutrition and Dietetics, Institute of Health Sciences, Gazi University, Ankara, Turkey
| | - Betül Sukan-Karaçağıl
- Department of Nutrition and Dietetics, Institute of Health Sciences, Gazi University, Ankara, Turkey
| | - Nilüfer Acar-Tek
- Faculty of Health Sciences, Department of Nutrition and Dietetics, Gazi University, Ankara, Turkey
| |
Collapse
|
45
|
Wang J, Liu W, Huang Y, Wang G, Guo X, Shi D, Sun T, Xiao C, Zhang C, Jiang B, Guo Y, Li J. A Senomorphlytic Three-Drug Combination Discovered in Salsola collina for Delaying Aging Phenotypes and Extending Healthspan. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401862. [PMID: 39073681 PMCID: PMC11423240 DOI: 10.1002/advs.202401862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 07/07/2024] [Indexed: 07/30/2024]
Abstract
The pursuit of pharmacological interventions in aging aims focuses on maximizing safety and efficacy, prompting an exploration of natural products endowed with inherent medicinal properties. Subsequently, this work establishes a unique library of plant extracts sourced from Yunnan Province, China. Screening of this herbal library herein revealed that Salsola collina (JM10001) notably enhances both lifespan and healthspan in C. elegans. Further analysis via network pharmacology indicates that the p53 signaling pathway plays a crucial role in mediating the anti-aging effects of JM10001. Additionally, this work identifies that a composition, designated as JM10101 and comprising three chemical constituents of JM10001, preserves the original lifespan-extending activity in C. elegans. Both JM10001 and JM10101 mitigate aging symptoms in senescence-accelerated mice treated with doxorubicin and in naturally aged mice. Notably, JM10101 exhibits a more sophisticated senomorphlytic role encompassing both senomorphic and senolytic functions than JM10001 in the modulation of senescent cells, offering a promising strategy for the discovery of combination drugs in the rational development of anti-aging therapies.
Collapse
Affiliation(s)
- Jiqun Wang
- State Key Laboratory of Bioreactor EngineeringShanghai Frontiers Science Center of Optogenetic Techniques for Cell MetabolismFrontiers Science Center for Materiobiology and Dynamic ChemistryShanghai Key Laboratory of New Drug DesignSchool of PharmacyEast China University of Science and TechnologyShanghai200237China
| | - Wenwen Liu
- Key Laboratory of Tropical Biological Resources of Ministry of EducationSchool of Pharmaceutical SciencesHainan UniversityHaikou570228China
| | - Yunyuan Huang
- Hubei Key Laboratory of Genetic Regulation and Integrative BiologySchool of Life SciencesCentral China Normal UniversityWuhanHubei430079China
| | - Guangwei Wang
- School of Chemical EngineeringKey Laboratory of Synthetic and Natural Functional Molecule of the Ministry of EducationNorthwest UniversityXi'an710127China
| | - Xiaobo Guo
- State Key Laboratory of Bioreactor EngineeringShanghai Frontiers Science Center of Optogenetic Techniques for Cell MetabolismFrontiers Science Center for Materiobiology and Dynamic ChemistryShanghai Key Laboratory of New Drug DesignSchool of PharmacyEast China University of Science and TechnologyShanghai200237China
| | - Donglei Shi
- Key Laboratory of Tropical Biological Resources of Ministry of EducationSchool of Pharmaceutical SciencesHainan UniversityHaikou570228China
| | - Tianyue Sun
- State Key Laboratory of Bioreactor EngineeringShanghai Frontiers Science Center of Optogenetic Techniques for Cell MetabolismFrontiers Science Center for Materiobiology and Dynamic ChemistryShanghai Key Laboratory of New Drug DesignSchool of PharmacyEast China University of Science and TechnologyShanghai200237China
| | - Chaojiang Xiao
- Yunnan Key Laboratory of Screening and Research on Anti‐pathogenic Plant Resources from Western Yunnan, Institute of Materia Medica & College of PharmacyDali UniversityDaliYunnan671000China
| | - Chao Zhang
- State Key Laboratory of Bioreactor EngineeringShanghai Frontiers Science Center of Optogenetic Techniques for Cell MetabolismFrontiers Science Center for Materiobiology and Dynamic ChemistryShanghai Key Laboratory of New Drug DesignSchool of PharmacyEast China University of Science and TechnologyShanghai200237China
| | - Bei Jiang
- Yunnan Key Laboratory of Screening and Research on Anti‐pathogenic Plant Resources from Western Yunnan, Institute of Materia Medica & College of PharmacyDali UniversityDaliYunnan671000China
| | - Yuan Guo
- School of Chemical EngineeringKey Laboratory of Synthetic and Natural Functional Molecule of the Ministry of EducationNorthwest UniversityXi'an710127China
| | - Jian Li
- State Key Laboratory of Bioreactor EngineeringShanghai Frontiers Science Center of Optogenetic Techniques for Cell MetabolismFrontiers Science Center for Materiobiology and Dynamic ChemistryShanghai Key Laboratory of New Drug DesignSchool of PharmacyEast China University of Science and TechnologyShanghai200237China
- Key Laboratory of Tropical Biological Resources of Ministry of EducationSchool of Pharmaceutical SciencesHainan UniversityHaikou570228China
- Key Laboratory of Xinjiang Phytomedicine Resource and UtilizationMinistry of EducationSchool of PharmacyShihezi UniversityShihezi832003China
| |
Collapse
|
46
|
Luo Y, Wen Y, Huang J, Chen B, Lv S, Qiu H, Li S, Liu S, Yang Q, He L, Yu Z, Zhao M, He M, Li D, Gu C. Matcha alleviates obesity by modulating gut microbiota and its metabolites. Curr Res Food Sci 2024; 9:100823. [PMID: 39253721 PMCID: PMC11381447 DOI: 10.1016/j.crfs.2024.100823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 08/02/2024] [Accepted: 08/13/2024] [Indexed: 09/11/2024] Open
Abstract
Matcha shows promise for diabetes, obesity, and gut microbiota disorders. Studies suggest a significant link between gut microbiota, metabolites, and obesity. Thus, matcha may have a positive impact on obesity by modulating gut microbiota and metabolites. This study used 16S rDNA sequencing and untargeted metabolomics to examine the cecal contents in mice. By correlation analysis, we explored the potential mechanisms responsible for the positive effects of matcha on obesity. The results indicated that matcha had a mitigating effect on the detrimental impacts of a high-fat diet (HFD) on multiple physiological indicators in mice, including body weight, adipose tissue weight, serum total cholesterol (TC), and low-density lipoprotein (LDL) levels, as well as glucose tolerance. Moreover, it was observed that matcha had an impact on the structural composition of gut microbiota and gut metabolites. Specifically, matcha was able to reverse the alterations in the abundance of certain obesity-improving bacteria, such as Alloprevotella, Ileibacterium, and Rikenella, as well as the abundance of obesity-promoting bacteria Romboutsia, induced by a HFD. Furthermore, matcha can influence the levels of metabolites, including formononetin, glutamic acid, pyroglutamic acid, and taurochenodeoxycholate, within the gastrointestinal tract. Additionally, matcha enhances caffeine metabolism and the HIF-1 signaling pathway in the KEGG pathway. The results of the correlation analysis suggest that formononetin, theobromine, 1,3,7-trimethyluric acid, and Vitamin C displayed negative correlation with both the obesity phenotype and microbiota known to exacerbate obesity, while demonstrating positive correlations with microbiota that alleviated obesity. However, glutamic acid, pyroglutamic acid, and taurochenodeoxycholate had the opposite effect. In conclusion, the impact of matcha on gut metabolites may be attributed to its modulation of the abundance of Alloprevotella, Ileibacterium, Rikenella, and Romboutsia within the gastrointestinal tract, thereby potentially contributing to the amelioration of obesity.
Collapse
Affiliation(s)
- Yadan Luo
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
- Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
| | - Yuhang Wen
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
- Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
| | - Jingrong Huang
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
- Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
| | - Baoting Chen
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
- Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
| | - Shuya Lv
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
- Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
| | - Hao Qiu
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
- Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
| | - Shuaibing Li
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
- School of Traditional Chinese Medicine, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, China
- Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
| | - Songwei Liu
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
- Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
| | - Qian Yang
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
- Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
| | - Lvqin He
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
- Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
| | - Zehui Yu
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
- Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
| | - Mingde Zhao
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
- Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
| | - Manli He
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
- Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
| | - Dong Li
- College of Bioengineering, Sichuan University of Science and Engineering, Yibin, 643002, China
| | - Congwei Gu
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
- Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
- Department of Nutrition and Food Hygiene, School of Public Health, Southwest Medical University, Luzhou, China
| |
Collapse
|
47
|
Zhao X, Ying J, Wang Z, Wang Y, Li Z, Gu T, Liu S, Li Y, Liu B, Xin F, Wen B. In vitro digestive properties and the bioactive effect of walnut green husk on human gut microbiota. Front Microbiol 2024; 15:1392774. [PMID: 39224223 PMCID: PMC11367867 DOI: 10.3389/fmicb.2024.1392774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 07/31/2024] [Indexed: 09/04/2024] Open
Abstract
Introduction Walnut green husk (WGH) is a waste byproduct from walnut industry. However, it is not well-known about its bioactive effect on human gut health. Methods This study conducted in vitro digestion and fermentation experiments to study the bioactive effect of WGH. Results Microbial fermentation was the primary mechanism to efficiently release phenolics and flavonoids, resulting in more excellent antioxidant capacities (DPPH, ABTS, and FRAP assays), which reached a highest value with 14.82 ± 0.01 mg VcE/g DW, 3.47 ± 0.01 mmol TE/g DW, and 0.96 ± 0.07 mmol FeSO4·7H2O/g DW, respectively. The surface microstructure of WGH became loose and fragmented after microbial fermentation. The analytical results of gut microbiota demonstrated that WGH could significantly increase the relative abundance of Proteobacteria in phylum level and Phascolarctobacterium in genus level while certain pro-inflammatory bacteria (such as Clostridium_sensu_stricto_1, Dorea, Alistipes, and Bilophila) was inhibited. Additionally, 1,373 differential metabolites were identified and enriched in 283 KEGG pathways. Of which some metabolites were significantly upregulated including ferulic acid, chlorogenic acid, umbelliferone, scopolin, muricholic acid, and so forth. Discussion These results indicated that WGH could have antioxidant and anti-inflammatory activities in the human gut, which could improve the economical value of WGH in the food industry.
Collapse
Affiliation(s)
- Xiaolan Zhao
- Laboratory of Biomanufacturing and Food Engineering, Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences (CAAS), Beijing, China
- Key Laboratory of Food Nutrition and Safety, College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin, China
| | - Jiabao Ying
- Laboratory of Biomanufacturing and Food Engineering, Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences (CAAS), Beijing, China
| | - Zhuochen Wang
- Institute of Agro-Products Processing, Anhui Academy of Agricultural Sciences, Hefei, China
| | - Yulu Wang
- Laboratory of Biomanufacturing and Food Engineering, Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences (CAAS), Beijing, China
| | - Zhen Li
- Laboratory of Biomanufacturing and Food Engineering, Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences (CAAS), Beijing, China
| | - Tianyi Gu
- Laboratory of Biomanufacturing and Food Engineering, Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences (CAAS), Beijing, China
| | - Shujun Liu
- Laboratory of Biomanufacturing and Food Engineering, Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences (CAAS), Beijing, China
| | - Yulong Li
- Laboratory of Biomanufacturing and Food Engineering, Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences (CAAS), Beijing, China
| | - Bing Liu
- Key Laboratory of Food Nutrition and Safety, College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin, China
| | - Fengjiao Xin
- Laboratory of Biomanufacturing and Food Engineering, Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences (CAAS), Beijing, China
- Laboratory of Biomanufacturing and Food Engineering, Institute of Agricultural Product Processing and Nutritional Health, Chinese Academy of Agricultural Sciences (CAAS), Cangzhou, China
| | - Boting Wen
- Laboratory of Biomanufacturing and Food Engineering, Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences (CAAS), Beijing, China
- Laboratory of Biomanufacturing and Food Engineering, Institute of Agricultural Product Processing and Nutritional Health, Chinese Academy of Agricultural Sciences (CAAS), Cangzhou, China
| |
Collapse
|
48
|
Wang K, Miao Y, Liu W, Muhammad I, Bao J, Jin X, Wu Z, Li R, Chen C, Li J. Lactobacillus salivarius ameliorates Mycoplasma gallisepticum-induced inflammation via the JAK/STAT signaling pathway involving respiratory microbiota and metabolites. Poult Sci 2024; 103:103942. [PMID: 38908119 PMCID: PMC11246048 DOI: 10.1016/j.psj.2024.103942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 05/20/2024] [Accepted: 05/30/2024] [Indexed: 06/24/2024] Open
Abstract
Mycoplasma gallisepticum (MG) can cause chronic respiratory disease (CRD) in chickens, which has a significant negative economic impact on the global poultry sector. Respiratory flora is the guardian of respiratory health, and its disorder is closely related to respiratory immunity and respiratory diseases. As a common probiotic in the chicken respiratory tract, Lactobacillus salivarius (L. salivarius) has potential antioxidant, growth performance enhancing, and anti-immunosuppressive properties. However, the specific mechanism through which L. salivarius protects against MG infection has not yet been thoroughly examined. This study intends to investigate whether L. salivarius could reduce MG-induced tracheal inflammation by modulating the respiratory microbiota and metabolites. The results indicated that L. salivarius reduced MG colonization significantly and alleviated the anomalous morphological changes by using the MG-infection model. L. salivarius also reduced the level of Th1 cell cytokines, increased the level of Th2 cell cytokines, and ameliorated immune imbalance during MG infection. In addition, L. salivarius improved the mucosal barrier, heightened immune function, and suppressed the Janus kinase/Signal transducer, and activator of transcription (JAK/STAT) signaling pathway. Notably, MG infection changed the composition of the respiratory microbiota and metabolites, and L. salivarius therapy partially reversed the aberrant respiratory microbiota and metabolite composition. Our results highlighted that these findings demonstrated that L. salivarius played a role in MG-mediated inflammatory damage and demonstrated that L. salivarius, by altering the respiratory microbiota and metabolites, could successfully prevent MG-induced inflammatory injury in chicken trachea.
Collapse
Affiliation(s)
- Kexin Wang
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Xiangfang District, Harbin 150030, P. R. China
| | - Yusong Miao
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Xiangfang District, Harbin 150030, P. R. China; Heilongjiang Academy of Agricultural Sciences, Harbin, 150086, P. R. China
| | - Weiqi Liu
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Xiangfang District, Harbin 150030, P. R. China
| | - Ishfaq Muhammad
- College of Computer Science, Huanggang Normal University, Huanggang, 438000, P. R. China
| | - Jiaxin Bao
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Xiangfang District, Harbin 150030, P. R. China
| | - Xiaodi Jin
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Xiangfang District, Harbin 150030, P. R. China
| | - Zhiyong Wu
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Xiangfang District, Harbin 150030, P. R. China
| | - Rui Li
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Xiangfang District, Harbin 150030, P. R. China
| | - Chunli Chen
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Xiangfang District, Harbin 150030, P. R. China; Department I of Pharmacology, Center for Pharmacology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, 50931, Germany
| | - Jichang Li
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Xiangfang District, Harbin 150030, P. R. China.
| |
Collapse
|
49
|
Liu Y, Wu H, Liu B, Chen S, Huang L, Liu Z, Wang J, Xie L, Wu X. Multi-omics analysis reveals the impact of gut microbiota on antipsychotic-induced weight gain in schizophrenia. Schizophr Res 2024; 270:325-338. [PMID: 38964078 DOI: 10.1016/j.schres.2024.06.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 06/16/2024] [Accepted: 06/22/2024] [Indexed: 07/06/2024]
Abstract
Emerging evidence indicates that gut microbial dysbiosis is associated with the development of antipsychotic-induced weight gain in schizophrenia (SZ). However, the exact taxonomic composition and functionality that constitute the "obesogenic" microbial profile remain elusive. Our retrospective survey identified two groups of the SZ population separated by BMI, with 1/3 of patients developing overweight/obesity after chronic antipsychotic treatment. Based on multi-omics analysis, we observed altered gut microbiota in SZ patients with overweight/obesity, characterized by a reduction in several beneficial bacteria genera, including Bacteroides, Parabacteroides, Akkermansia, and Clostridium. This microbial dysbiosis was accompanied by disrupted energy expenditure and nutritional metabolism, worsened metabolic indices, and reduced levels of beneficial metabolites, e.g. indole-3-carboxylic acid and propionic acid. Moreover, leveraging data from first-episode drug-naïve schizophrenia (FSZ) patients at one-month and one-year follow-up, both artificial neural network and random forest classifier-based prediction models demonstrated a strong ability of microbial profiles to predict antipsychotic-induced weight gain. Importantly, FSZ patients with higher relative abundance of Parabacteria distasonis were less susceptible to antipsychotic-induced weight gain. Thus, gut microbiota could serve as a noninvasive approach to predict antipsychotic-induced weight gain, guiding clinical antipsychotics administration and developing novel therapeutic strategies for weight management in SZ.
Collapse
Affiliation(s)
- Yaxi Liu
- Psychiatry Department, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510630, China; Sleep Medicine Center of Psychiatry Department, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Hui Wu
- Radiology Department, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Bingdong Liu
- Department of Endocrinology and Metabolism, Zhujiang Hospital of Southern Medical University, Guangzhou 510280, China; State Key Laboratory of Applied Microbiology Southern China, Guangdong Institute of Microbiology, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China
| | - Shengyun Chen
- Psychiatry Department, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510630, China
| | - Liujing Huang
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Institute of Microbiology, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China
| | - Zhihong Liu
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Institute of Microbiology, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China
| | - Jie Wang
- Department of Life Sciences, Imperial College London, London SW7 2AZ, United Kingdom
| | - Liwei Xie
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Institute of Microbiology, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China.
| | - Xiaoli Wu
- Psychiatry Department, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510630, China.
| |
Collapse
|
50
|
Chen C, Feng F, Qi M, Chen Q, Tang W, Diao H, Hu Z, Qiu Y, Li Z, Chu Y, Tang Z. Dietary Citrus Flavonoids Improved Growth Performance and Intestinal Microbiota of Weaned Piglets via Immune Function Mediated by TLR2/NF-κB Signaling Pathway. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:16761-16776. [PMID: 39012185 DOI: 10.1021/acs.jafc.4c03401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/17/2024]
Abstract
This study investigated the effects of citrus flavonoids (CF) and compared to antibiotics on piglet growth and gut health. Weaned piglets were fed either a basal diet (CON) or a basal diet supplemented with 75 mg/kg chlortetracycline (CTC), 20 mg/kg CF (CF1), 40 mg/kg CF (CF2), or 80 mg/kg CF (CF3). The CF group, especially CF3, exhibited improved growth performance; reduced diarrhea; significantly higher levels of serum growth factors, immunoglobulins, and anti-inflammatory cytokines; and significantly lower levels of pro-inflammatory factors and markers of intestinal barrier damage (P < 0.05). The intestinal mucosa proteins ZO-1 and occludin increased, while NF-κB and TLR2 decreased, correlating with CF dosage (P < 0.05). Furthermore, CF promoted a favorable balance in the gut microbiota, with an increased relative abundance of Bacteroidetes and Prevotella and decreased taxa Tenericutes and Clostridiales. Overall, CF enhanced piglet growth and gut health by modulating the TLR2/NF-κB pathway, offering a natural antibiotic alternative. The optimal dose of CF was 80 mg/kg.
Collapse
Affiliation(s)
- Chen Chen
- Animal Nutrition and Bio-feed, Chongqing Key Laboratory of Herbivore Science, College of Animal Science and Technology, Southwest University, Chongqing 400715, China
| | - Fu Feng
- Animal Nutrition and Bio-feed, Chongqing Key Laboratory of Herbivore Science, College of Animal Science and Technology, Southwest University, Chongqing 400715, China
| | - Min Qi
- Yunnan Animal Husbandry Station, Kunming 650225, China
| | - Qingju Chen
- Animal Nutrition and Bio-feed, Chongqing Key Laboratory of Herbivore Science, College of Animal Science and Technology, Southwest University, Chongqing 400715, China
| | - Wenjie Tang
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China
- Livestock and Poultry Biological Products Key Laboratory of Sichuan Province, Sichuan Animtche Group Co., Ltd., Chengdu 610066, China
| | - Hui Diao
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China
- Livestock and Poultry Biological Products Key Laboratory of Sichuan Province, Sichuan Animtche Group Co., Ltd., Chengdu 610066, China
| | - Zhijin Hu
- Animal Nutrition and Bio-feed, Chongqing Key Laboratory of Herbivore Science, College of Animal Science and Technology, Southwest University, Chongqing 400715, China
| | - Yibin Qiu
- Animal Nutrition and Bio-feed, Chongqing Key Laboratory of Herbivore Science, College of Animal Science and Technology, Southwest University, Chongqing 400715, China
| | - Zhangcheng Li
- Animal Nutrition and Bio-feed, Chongqing Key Laboratory of Herbivore Science, College of Animal Science and Technology, Southwest University, Chongqing 400715, China
| | - Yunyun Chu
- Animal Nutrition and Bio-feed, Chongqing Key Laboratory of Herbivore Science, College of Animal Science and Technology, Southwest University, Chongqing 400715, China
| | - Zhiru Tang
- Animal Nutrition and Bio-feed, Chongqing Key Laboratory of Herbivore Science, College of Animal Science and Technology, Southwest University, Chongqing 400715, China
| |
Collapse
|