1
|
Tuersong T, Wu QF, Chen Y, Shan Li P, Yong YX, Shataer M, Shataer S, Ma LY, Yang XL. Integrated network pharmacology, metabolomics, and microbiome studies to reveal the therapeutic effects of Anacyclus pyrethrum in PD-MCI mice. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 142:156729. [PMID: 40253741 DOI: 10.1016/j.phymed.2025.156729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 03/03/2025] [Accepted: 04/02/2025] [Indexed: 04/22/2025]
Abstract
BACKGROUND Anacyclus pyrethrum (l.) DC has potential value in treating Parkinson's disease (PD)-mild cognitive impairment (MCI), manifesting as impaired memory, attention, executive function, and language. However, the specific targets and modes of action of A. pyrethrum remain unclear. PURPOSE The aim of this study was to identify the active components of A. pyrethrum and examine their effectiveness in treating a mouse model of PD-MCI. METHODS We generated ethanol extracts of A. pyrethrum root (EEAP) and identified its active components and related targets using UHPLC-MS/MS and network pharmacology.The PD-MCI model was induced via intraperitoneal administration of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine(MPTP). After following continuous administration of EEAP,Altered learning or memory, as well as anxiety, were tested using the morris water maze, eight-arm radial arm maze (RAM), and open-field test,elevated plus-maze. Brain histopathology and ultrastructural changes were examined using brightfield microscopy, and electron microscopy, respectively. Furthermore, protein expression was assessed using western blotting.Stool samples were used for metabolomics analysis by UHPLC-MS/MS and for 16S rDNA sequencing to determine the compositional changes of the gut microbiota.We conducted a short-chain fatty acid targeted metabolomics experiment to study their role in the gut-brain axis in PD-MCI. RESULTS Using UPLC-MS-MS, 126 compounds were identified from A. pyrethrum samples.After searching the databases and literature reports, 31 active components and 544 drug-disease targets were screened. Biological processes and molecular functions, such as energy channels, cell signaling, and metabolism, were discovered through GO analysis. The water maze experiment showed that the average swimming distance and escape latency of mice in EEAP groups decreased. The eight-arm maze experiment showed that model had a much higher number of errors related to working memory than the control mice. In the open field experiment, compared with the control group, the mice in the EEAP group exhibited an increase in the average movement speed and total movement distance, along with a decrease in the residence time.In the elevated plus maze, the control had less anxiety than the Model. Donepezil/Levodopa(D/l) mitigated anxiety-like behavior, and EEAP (100-400 mg/kg) showed a dose-dependent increase in open-arm metrics, suggesting it may ease anxiety in mice.Hippocampal tissue of mice treated with different doses of EEAP showed intact cellular layers and the hematoxylin-eosin-stained cones were slightly better;cells were arranged neatly; their morphology was normal, and were distributed uniformly. Electron microscopy revealed that the nuclear membrane, chromatin, and nucleoli were clearly demarcated in the hippocampus of mice treated with different doses of EEAP, contrary to that in the model group. In brain extracts of the EEAP group, lighter thinner bands for amyloid precursor protein (APP) and Aβ were observed compared to those in the model group. In model mice, APP and Aβ protein expression was higher than in the blank group, as shown by stronger bands. In EEAP-treated mice, the bands were weaker, indicating reduced expression. In the model group had lower Bcl-2 and higher Bax levels. EEAP treatment increased Bcl-2 and decreased Bax expression.Compared to the control group, the model showed substantially low glutathione peroxidase (GSH-Px),superoxide dismutase(SOD),catalase (CAT)activity (p < 0.05),much higher (p < 0.05) in the EEAP-H group than that in the model. EEAP intervention significantly modulated the fecal metabolic profile of PD-MCI mice. The abundance of steroid and lipid metabolites, including linoleylethanolamine, was markedly altered in the model group compared to the control group, with EEAP treatment reversing several of these abnormalities. PLS-DA and OPLS-DA revealed significant separation between groups (Q2= 0.542, p < 0.01), confirming a dose-dependent effect. Random forest analysis identified 15 key metabolic markers, such as dose-dependent changes in d-glutamine and hydrocodone. Metabolic pathway analysis demonstrated significant enrichment in phenylalanine, tyrosine, tryptophan metabolism, and arginine biosynthesis pathways (p < 0.05). The Support Vector Machine (SVM) model achieved an AUC approaching 1, indicating substantial differences in metabolite profiles. EEAP intervention significantly influenced the composition and functional profile of the intestinal microbiota. The Venn diagram illustrates that each group shared 342 operational taxonomic units (OTUs), with the EEAP 400 group exhibiting a distinct Bacteroidetes proportion. LEfSe analysis identified g_Prevotella as the characteristic bacterium in the control group, c_Epsilonproteobacteria in the model group, and g_Adlercreutzia in the EEAP 100 group. The Faith's Phylogenetic Diversity (PD) index was highest in the EEAP 100 group, and Non-metric Multidimensional Scaling (NMDS)/Principal Coordinates Analysis (PCoA) revealed significant differences in microbial community structure. Short-chain fatty acids (SCFAs) analysis indicated that acetic acid was the predominant metabolite, while EEAP dose-dependently regulated propionic acid and isovaleric acid levels (VIP > 1, p < 0.001). These findings demonstrate that EEAP exerts its regulatory effects by reshaping the structure and metabolic functions of the gut microbiota. CONCLUSION EEAP holds great promise as a potential therapeutic agent for PD-MCI, exerting its effects through multiple mechanisms, including regulating protein expression, modulating the fecal metabolic profile, and reshaping the gut microbiota and its metabolites.
Collapse
Affiliation(s)
- Tayier Tuersong
- Department of Pharmacy, Xinjiang Key Laboratory of Neurological Diseases, Xinjiang Clinical Research Center for Nervous System Diseases, Second Affiliated Hospital of Xinjiang Medical University, Ürümqi 830001, Xinjiang, PR China
| | - Qin Fen Wu
- Department of Neurology, Xinjiang Key Laboratory of Neurological Diseases, Xinjiang Clinical Research Center for Nervous System Diseases, Second Affiliated Hospital of Xinjiang Medical University, Ürümqi 830001, Xinjiang, PR China
| | - Yan Chen
- Department of Pharmacy, Xinjiang Key Laboratory of Neurological Diseases, Xinjiang Clinical Research Center for Nervous System Diseases, Second Affiliated Hospital of Xinjiang Medical University, Ürümqi 830001, Xinjiang, PR China
| | - Pei Shan Li
- Department of Neurology, Xinjiang Key Laboratory of Neurological Diseases, Xinjiang Clinical Research Center for Nervous System Diseases, Second Affiliated Hospital of Xinjiang Medical University, Ürümqi 830001, Xinjiang, PR China
| | - Yu Xuan Yong
- Department of Neurology, Xinjiang Key Laboratory of Neurological Diseases, Xinjiang Clinical Research Center for Nervous System Diseases, Second Affiliated Hospital of Xinjiang Medical University, Ürümqi 830001, Xinjiang, PR China
| | - Munire Shataer
- Department of Histology and Embryology, Basic Medical College of Xinjiang Medical University, Ürümqi 830001, Xinjiang, PR China
| | - Samire Shataer
- Department of Neurology, Xinjiang Key Laboratory of Neurological Diseases, Xinjiang Clinical Research Center for Nervous System Diseases, Second Affiliated Hospital of Xinjiang Medical University, Ürümqi 830001, Xinjiang, PR China
| | - Liang Ying Ma
- Department of Pharmacy, Xinjiang Key Laboratory of Neurological Diseases, Xinjiang Clinical Research Center for Nervous System Diseases, Second Affiliated Hospital of Xinjiang Medical University, Ürümqi 830001, Xinjiang, PR China
| | - Xin Ling Yang
- Department of Neurology, Xinjiang Key Laboratory of Neurological Diseases, Xinjiang Clinical Research Center for Nervous System Diseases, Second Affiliated Hospital of Xinjiang Medical University, Ürümqi 830001, Xinjiang, PR China.
| |
Collapse
|
2
|
Yang J, Wang X, Chen Y, He Y, Li P, Wen X, Wang B. ChangQing compound relieves Eimeria tenella infection symptoms by modulating intestinal probiotic and pathogenic bacteria balance. Vet J 2025; 311:106343. [PMID: 40187631 DOI: 10.1016/j.tvjl.2025.106343] [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: 10/22/2024] [Revised: 03/30/2025] [Accepted: 03/31/2025] [Indexed: 04/07/2025]
Abstract
Cecal coccidiosis is a severe and lethal parasitic disease affecting chickens, making the search for effective preventive agents free of contamination and drug resistance crucial for controlling this condition in poultry. Previous studies have demonstrated that the ChangQing compound has significant therapeutic effects against cecal coccidiosis; however, its potential as a preventive measure has yet to be evaluated. In this study, we established an experimental model for the prevention of cecal coccidiosis in chickens using the ChangQing compound for the first time. A comprehensive evaluation was performed on survival rates, relative weight gain, oocyst production, the anticoccidial index (ACI), immune parameters, parasitic tissue pathology, and microbial diversity in cecal contents. Results indicated that the ChangQing compound at a concentration of 5.0 g/L achieved an ACI of 178.10 in the Pre-Exposure Prophylaxis (PrEP) group and 173.12 in the Post-Exposure Prophylaxis (PEP) group. Compared to the positive control group, cecal lesions were reduced, and indices for the spleen, liver, and bursa of Fabricius increased in both experimental groups following ChangQing administration. Furthermore, levels of immune factors, IgA, IgG, and IgM significantly elevated. The abundance of beneficial bacteria, including Lactobacillus, Bacteroides, and Alistipes, increased in the 5.0 g/L ChangQing compound group, while potential pathogens like Escherichia-Shigella, Enterococcus, and norank_f_Oscillospiraceae were reduced. These findings offer critical data for coccidiosis prevention in chickens and lay a theoretical foundation for future research on the antiparasitic mechanisms of traditional Chinese medicine.
Collapse
Affiliation(s)
- Jiajia Yang
- College of Animal Science, Guizhou University, Guiyang, Guizhou Province 550025, PR China; Key Laboratory of Animal Diseases and Veterinary Public Health of Guizhou Province (Cultivation), Guiyang, Guizhou Province 550025, PR China.
| | - Xuan Wang
- Institute of Animal Husbandry and Veterinary Medicine of Guizhou Academy of Agricultural Sciences, Guizhou Province 550025, PR China.
| | - Ying Chen
- College of Animal Science, Guizhou University, Guiyang, Guizhou Province 550025, PR China; Key Laboratory of Animal Diseases and Veterinary Public Health of Guizhou Province (Cultivation), Guiyang, Guizhou Province 550025, PR China.
| | - Ye He
- College of Animal Science, Guizhou University, Guiyang, Guizhou Province 550025, PR China; Key Laboratory of Animal Diseases and Veterinary Public Health of Guizhou Province (Cultivation), Guiyang, Guizhou Province 550025, PR China.
| | - Ping Li
- College of Animal Science, Guizhou University, Guiyang, Guizhou Province 550025, PR China; Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang, Guizhou Province 550025, PR China.
| | - Xin Wen
- College of Animal Science, Guizhou University, Guiyang, Guizhou Province 550025, PR China; Key Laboratory of Animal Diseases and Veterinary Public Health of Guizhou Province (Cultivation), Guiyang, Guizhou Province 550025, PR China; Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang, Guizhou Province 550025, PR China.
| | - Bi Wang
- College of Animal Science, Guizhou University, Guiyang, Guizhou Province 550025, PR China; Key Laboratory of Animal Diseases and Veterinary Public Health of Guizhou Province (Cultivation), Guiyang, Guizhou Province 550025, PR China; Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang, Guizhou Province 550025, PR China.
| |
Collapse
|
3
|
Zhang J, Shen M, Yin Y, Chen Y, Deng X, Mo J, Zhou X, Lin J, Chen X, Xie X, Wu X, Chen X. Carnosic acid reduces lipid content, enhances gut health, and modulates microbiota composition and metabolism in diet-induced obese mice. Food Funct 2025; 16:1888-1902. [PMID: 39932492 DOI: 10.1039/d4fo04534c] [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: 03/04/2025]
Abstract
Carnosic acid (CA) is a bioactive phenolic diterperne compound found in sage and rosemary. The present study investigated the beneficial effects of CA (50 and 100 mg per kg bw) in diet-induced obese mice and the underlying mechanisms of action. After the intervention, the physiology, lipid metabolism, and tissue morphology, as well as the inflammation, gut microbiota, and metabolomics in the colon were measured. We found that CA improved the composition and metabolism of the gut microbiota in obese mice, with Akkermansia being the dominant bacterium negatively correlated with obesity and various fecal metabolites. Regarding the intestinal barrier function, CA promoted the expression of tight junction proteins and inhibited the TLR4/MyD88/NF-κB signaling pathway in obese mice to alleviate colonic inflammation. These results suggest that CA improved multiple aspects of gut health in diet-induced obesity in mice, providing a scientific basis for future clinical studies in humans.
Collapse
Affiliation(s)
- Jing Zhang
- School of Public Health, Guangzhou Medical University, Guangzhou 510642, Guangdong, P. R. China.
| | - Mengzhu Shen
- School of Public Health, Guangzhou Medical University, Guangzhou 510642, Guangdong, P. R. China.
| | - Yue Yin
- School of Public Health, Guangzhou Medical University, Guangzhou 510642, Guangdong, P. R. China.
| | - Yuru Chen
- School of Public Health, Guangzhou Medical University, Guangzhou 510642, Guangdong, P. R. China.
| | - Xianying Deng
- School of Public Health, Guangzhou Medical University, Guangzhou 510642, Guangdong, P. R. China.
| | - Jingyun Mo
- School of Public Health, Guangzhou Medical University, Guangzhou 510642, Guangdong, P. R. China.
| | - Xiaoling Zhou
- School of Public Health, Guangzhou Medical University, Guangzhou 510642, Guangdong, P. R. China.
| | - Juanying Lin
- School of Public Health, Guangzhou Medical University, Guangzhou 510642, Guangdong, P. R. China.
| | - Xinxin Chen
- School of Public Health, Guangzhou Medical University, Guangzhou 510642, Guangdong, P. R. China.
| | - Xinwei Xie
- School of Public Health, Guangzhou Medical University, Guangzhou 510642, Guangdong, P. R. China.
| | - Xian Wu
- Department of Kinesiology, Nutrition, and Health, Miami University, Oxford, Ohio 45056, USA.
| | - Xuexiang Chen
- School of Public Health, Guangzhou Medical University, Guangzhou 510642, Guangdong, P. R. China.
| |
Collapse
|
4
|
Wang M, Yang T, Xiang Y, Pang J, Wang Y, Sun D. Coix Seed Extract Attenuates Glycolipid Metabolism Disorder in Hyperlipidemia Mice Through PPAR Signaling Pathway Based on Metabolomics and Network Pharmacology. Foods 2025; 14:770. [PMID: 40077474 PMCID: PMC11899454 DOI: 10.3390/foods14050770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Revised: 02/19/2025] [Accepted: 02/20/2025] [Indexed: 03/14/2025] Open
Abstract
Hyperlipidemia is characterized by a high level of blood lipid which poses a serious threat to human health. Coix seed is a traditional crop of medicine and food homology with a wide range of pharmacological actions. To make clear the attenuation effect of coix seed against hyperlipidemia, low and high doses of coix seed extract (CSE) were orally administered to hyperlipidemia model mice developed by high-fat diet (HFD). Our results showed that CSE notably improved liver pathological injury, and oxidative stress, and declined the levels of glucose and lipid in hyperlipidemia mice. Liver metabolomics showed that lipid-related metabolites notably decreased, and pathways of glycolipid metabolism were seriously affected by CSE intervention. Moreover, 16S rRNA sequencing revealed that CSE treatment notably increased the diversity of gut microbiota. Meanwhile, the microbiota with the function of regulating intestinal balance as well as relieving obesity and nervous diseases significantly enhanced while harmful flora notably decreased after CSE intervention. The results of network pharmacology and molecular docking indicated that the PPAR signaling pathway may be the core path of anti-hyperlipidemia for coix seeds. RT-qPCR further verified that the expression levels of genes from the PPAR pathway notably changed by CSE treatment with fat synthesis genes significantly decreased while lipolysis genes notably enhanced. Therefore, coix seed might be a potential candidate for the treatment of hyperlipidemia.
Collapse
Affiliation(s)
- Min Wang
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang 561113, China; (M.W.); (T.Y.); (Y.X.); (Y.W.)
| | - Tianming Yang
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang 561113, China; (M.W.); (T.Y.); (Y.X.); (Y.W.)
| | - Yongjing Xiang
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang 561113, China; (M.W.); (T.Y.); (Y.X.); (Y.W.)
| | - Junxiao Pang
- College of Food Science and Engineering, Guiyang University, Guiyang 550005, China;
| | - Yao Wang
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang 561113, China; (M.W.); (T.Y.); (Y.X.); (Y.W.)
| | - Dali Sun
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang 561113, China; (M.W.); (T.Y.); (Y.X.); (Y.W.)
| |
Collapse
|
5
|
Feng BY, Chen PL, Yan L, Huang WF, Li CF, Yi LT, Xu GH. Long-term Pu-erh tea alleviates inflammatory bowel disease via the regulation of intestinal microbiota and maintaining the intestinal mucosal barrier. Food Sci Biotechnol 2025; 34:743-755. [PMID: 39958166 PMCID: PMC11822139 DOI: 10.1007/s10068-024-01696-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 08/12/2024] [Accepted: 08/19/2024] [Indexed: 02/18/2025] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic gastrointestinal condition with increasing global prevalence. Current therapies are limited, leading to exploration of novel treatments like Pu-erh tea, a fermented tea recognized for its health benefits. This study shows that long-term consumption of Pu-erh tea significantly reduces IBD symptoms in DSS-induced mice by moderating inflammation and enhancing oxidative responses in the colon. Pu-erh tea notably increases the abundance of specific gut microbiota, particularly enhancing Firmicutes, Bacteroidota, and Proteobacteria phyla, and raising levels of Lactobacillus and Muribaculaceae genera. Key species such as Lactobacillus johnsonii, Lactobacillus reuteri, and Lactobacillus murinus also showed increased abundance. Additionally, Pu-erh tea helps restore the integrity of the intestinal barrier. These findings highlight the potential of Pu-erh tea as a complementary dietary strategy for IBD, potentially improving disease management and patient outcomes through its effects on the intestinal microbiota and mucosal barrier. Supplementary Information The online version contains supplementary material available at 10.1007/s10068-024-01696-9.
Collapse
Affiliation(s)
- Bi-Yun Feng
- Fujian University of Traditional Chinese Medicine College of Pharmacy, Fuzhou, 350108 Fujian People’s Republic of China
- Xiamen Medicine Research Institute, Xiamen, 361008 Fujian People’s Republic of China
| | - Pei-Lu Chen
- Fujian University of Traditional Chinese Medicine College of Pharmacy, Fuzhou, 350108 Fujian People’s Republic of China
- Xiamen Medicine Research Institute, Xiamen, 361008 Fujian People’s Republic of China
| | - Ling Yan
- Fujian University of Traditional Chinese Medicine College of Pharmacy, Fuzhou, 350108 Fujian People’s Republic of China
- Xiamen Medicine Research Institute, Xiamen, 361008 Fujian People’s Republic of China
| | - Wei-Feng Huang
- Department of Gastroenterology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003 Fujian People’s Republic of China
| | - Cheng-Fu Li
- Xiamen Hospital of Traditional Chinese Medicine, Xiamen, 361009 Fujian People’s Republic of China
| | - Li-Tao Yi
- Department of Chemical and Pharmaceutical Engineering, College of Chemical Engineering, Huaqiao University, Xiamen, 361021 Fujian People’s Republic of China
- Institute of Pharmaceutical Engineering, Huaqiao University, Xiamen, 361021 Fujian People’s Republic of China
- Fujian Provincial Key Laboratory of Biochemical Technology, Huaqiao University, Xiamen, 361021 Fujian People’s Republic of China
| | - Guang-Hui Xu
- Fujian University of Traditional Chinese Medicine College of Pharmacy, Fuzhou, 350108 Fujian People’s Republic of China
- Xiamen Medicine Research Institute, Xiamen, 361008 Fujian People’s Republic of China
- Xiamen Key Laboratory of Natural Medicine Research and Development, Xiamen, 361021 Fujian People’s Republic of China
| |
Collapse
|
6
|
Chen N, Yao P, Farid MS, Zhang T, Luo Y, Zhao C. Effect of bioactive compounds in processed Camellia sinensis tea on the intestinal barrier. Food Res Int 2025; 199:115383. [PMID: 39658174 DOI: 10.1016/j.foodres.2024.115383] [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: 08/17/2024] [Revised: 10/18/2024] [Accepted: 11/14/2024] [Indexed: 12/12/2024]
Abstract
The human intestinal tract plays a pivotal role in safeguarding the body against noxious substances and microbial pathogens by functioning as a barrier. This barrier function is achieved through the combined action of physical, chemical, microbial, and immune components. Tea (Camellia sinensis) is the most widely consumed beverage in the world, and it is consumed and appreciated in a multitude of regions across the globe. Tea can be classified into various categories, including green, white, yellow, oolong, black, and dark teas, based on the specific processing methods employed. In recent times, there has been a notable surge in scientific investigation into the various types of tea. The recent surge in research on tea can be attributed to the plethora of bioactive compounds it contains, including polyphenols, polysaccharides, pigments, and theanine. The processing of different teas affects the active ingredients to varying degrees, resulting in a range of chemical reactions and the formation of different types and quantities of ingredients. The bioactive compounds present in tea are of great importance for the maintenance of the integrity of the intestinal barrier, operating through a variety of mechanisms. This literature review synthesizes scientific studies on the impact of the primary bioactive compounds and different processing methods of tea on the intestinal barrier function. This review places particular emphasis on the exploration of the barrier repair and regulatory effects of these compounds, including the mitigation of damage to different barriers following intestinal diseases. Specifically, the active ingredients in tea can alleviate damage to physical barriers and chemical barriers by regulating barrier protein expression. At the same time, they can also maintain the stability of immune and biological barriers by regulating the expression of inflammatory factors and the metabolism of intestinal flora. This investigation can establish a strong theoretical foundation for the future development of innovative tea products.
Collapse
Affiliation(s)
- Nan Chen
- College of Food Science and Engineering, Jilin University, Changchun 130062, China.
| | - Peng Yao
- College of Food Science and Engineering, Jilin University, Changchun 130062, China.
| | | | - Tiehua Zhang
- College of Food Science and Engineering, Jilin University, Changchun 130062, China.
| | - Yangchao Luo
- Department of Nutritional Sciences, University of Connecticut, Storrs, CT 06269, United States.
| | - Changhui Zhao
- College of Food Science and Engineering, Jilin University, Changchun 130062, China.
| |
Collapse
|
7
|
Wang C, Qiu M, Wang S, Luo J, Huang L, Deng Q, Fang Z, Sun L, Gooneratne R. Gut-Microbiota-Derived Butyric Acid Overload Contributes to Ileal Mucosal Barrier Damage in Late Phase of Chronic Unpredictable Mild Stress Mice. Int J Mol Sci 2024; 25:12998. [PMID: 39684708 DOI: 10.3390/ijms252312998] [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: 10/19/2024] [Revised: 11/24/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
Intestinal mucosal barrier damage is regarded as the critical factor through which chronic unpredictable mild stress (CUMS) leads to a variety of physical and mental health problems. However, the exact mechanism by which CUMS induces intestinal mucosal barrier damage is unclear. In this study, 14, 28, and 42 d CUMS model mice were established. The indicators related to ileal mucosal barrier damage (IMBD), the composition of the ileal microbiota and its amino acid (AA) and short-chain fatty acid (SCFA) metabolic functions, and free amino acid (FAA) and SCFA levels in the ileal lumen were measured before and after each stress period. The correlations between them are analyzed to investigate how CUMS induces intestinal mucosal barrier damage in male C57BL/6 mice. With the progression of CUMS, butyric acid (BA) levels decreased (14 and 28 d) and then increased (42 d), and IMBD progressively increased. In the late CUMS stage (42 d), the degree of IMBD is most severe and positively correlated with significantly increased BA levels (p < 0.05) in the ileal lumen and negatively correlated with significantly decreased FAAs, such as aspartic, glutamic, alanine, and glycine levels (p < 0.05). In the ileal lumen, the abundance of BA-producing bacteria (Muribaculaceae, Ruminococcus, and Butyricicoccus) and the gene abundance of specific AA degradation and BA production pathways and their related enzymes are significantly increased (p < 0.05). In addition, there is a significant decrease (p < 0.05) in the abundance of core bacteria (Prevotella, Lactobacillus, Turicibacter, Blautia, and Barnesiella) that rely on these specific AAs for growth and/or are sensitive to BA. These changes, in turn, promote further colonization of BA-producing bacteria, exacerbating the over-accumulation of BA in the ileal lumen. These results were validated by ileal microbiota in vitro culture experiments. In summary, in the late CUMS stages, IMBD is related to an excessive accumulation of BA caused by dysbiosis of the ileal microbiota and its overactive AA degradation.
Collapse
Affiliation(s)
- Chen Wang
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China
| | - Mei Qiu
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China
| | - Shuo Wang
- College of Agriculture and Biotechnology, Sun Yat-sen University, Shenzhen 518107, China
| | - Jinjin Luo
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China
| | - Ling Huang
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China
| | - Qi Deng
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China
| | - Zhijia Fang
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China
| | - Lijun Sun
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China
| | - Ravi Gooneratne
- Department of Wine, Food and Molecular Biosciences, Faculty of Agriculture and Life Sciences, Lincoln University, P.O. Box 85084, Lincoln 7647, New Zealand
| |
Collapse
|
8
|
Liang C, Wei S, Ji Y, Lin J, Jiao W, Li Z, Yan F, Jing X. The role of enteric nervous system and GDNF in depression: Conversation between the brain and the gut. Neurosci Biobehav Rev 2024; 167:105931. [PMID: 39447778 DOI: 10.1016/j.neubiorev.2024.105931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 10/14/2024] [Accepted: 10/20/2024] [Indexed: 10/26/2024]
Abstract
Depression is a debilitating mental disorder that causes a persistent feeling of sadness and loss of interest. Approximately 280 million individuals worldwide suffer from depression by 2023. Despite the heavy medical and social burden imposed by depression, pathophysiology remains incompletely understood. Emerging evidence indicates various bidirectional interplay enable communication between the gut and brain. These interplays provide a link between intestinal and central nervous system as well as feedback from cortical and sensory centers to enteric activities, which also influences physiology and behavior in depression. This review aims to overview the significant role of the enteric nervous system (ENS) in the pathophysiology of depression and gut-brain axis's contribution to depressive disorders. Additionally, we explore the alterations in enteric glia cells (EGCs) and glial cell line-derived neurotrophic factor (GDNF) in depression and their involvement in neuronal support, intestinal homeostasis maintains and immune response activation. Modulating ENS function, EGCs and GDNF level could serve as novel strategies for future antidepressant therapy.
Collapse
Affiliation(s)
- Chuoyi Liang
- School of Nursing, Jinan University, Guangzhou, China
| | - Sijia Wei
- School of Nursing, Jinan University, Guangzhou, China
| | - Yelin Ji
- School of Nursing, Jinan University, Guangzhou, China
| | - Jiayi Lin
- School of Nursing, Jinan University, Guangzhou, China
| | - Wenli Jiao
- School of Nursing, Jinan University, Guangzhou, China
| | - Zhiying Li
- School of Nursing, Jinan University, Guangzhou, China
| | - Fengxia Yan
- School of Nursing, Jinan University, Guangzhou, China.
| | - Xi Jing
- School of Nursing, Jinan University, Guangzhou, China; Guangdong-Hong Kong-Macau Great Bay Area Geoscience Joint Laboratory, School of Medicine, Jinan University, Guangzhou, China.
| |
Collapse
|
9
|
Chen X, Wu J, Zhou B, Zhu M, Zhang J, Zhou N, Zhu YZ, Zhang X, Duan X, Men K. Bacterial Lysate-Based Bifunctional mRNA Nanoformulation for Efficient Colon Cancer Immunogene Therapy. ACS APPLIED MATERIALS & INTERFACES 2024; 16:56580-56598. [PMID: 39397736 DOI: 10.1021/acsami.4c07684] [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: 10/15/2024]
Abstract
mRNA-based nonviral gene therapy has played an important role in cancer therapy, however, the limited delivery efficiency and therapeutic capacity still require further exploration and enhancement. Immunogene therapy provides a strategy for cancer treatment. Bacteria are tiny single-celled living organisms, many of which can be found in and on the human body and are beneficial to humans. Lactobacillus reuteri is a bacterial member of the gut flora, and recent research has shown that it can reduce intestinal inflammation by stimulating an immunomodulatory response. L. reuteri lysate represents an ideal resource for constructing advanced mRNA delivery systems with immune stimulation potential. Here, we prepared a bifunctional mRNA delivery system DMP-Lac (DOTAP-mPEG-PCL-L. reuteri lysate), which successfully codelivered L. reuteri lysate and IL-23A mRNA, exhibited a high mRNA delivery efficiency of 75.56% ± 0.85%, and strongly promoted the maturation and activation of the immune system in vivo. Both the CT26 abdominal metastasis model and the lung metastasis model also exhibited a good therapeutic effect, and the tumor inhibition rate of DMP-Lac/IL-23A group reached 97.92%. Protein chip technology verified that DMP acted as an immune adjuvant, demonstrating that the L. reuteri lysate could regulate the related immune cells, while IL-23 mRNA caused changes in downstream factors, thus producing the corresponding tumor treatment effect. The DMP-Lac/IL-23A complex exhibited strong anticancer immunotherapeutic effects. Our results demonstrated that this bifunctional mRNA formulation served as a tumor-specific nanomedicine, providing an advanced strategy for colon cancer immunogene therapy.
Collapse
Affiliation(s)
- Xiaohua Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China
| | - Jieping Wu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China
| | - Bailing Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China
| | - Manfang Zhu
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Jin Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China
| | - Na Zhou
- State Key Laboratory for Quality Research of Chinese Medicines and School of Pharmacy, Macau University of Science and Technology, Taipa, Macau 999078, China
| | - Yi Zhun Zhu
- State Key Laboratory for Quality Research of Chinese Medicines and School of Pharmacy, Macau University of Science and Technology, Taipa, Macau 999078, China
| | - Xin Zhang
- State Key Laboratory for Quality Research of Chinese Medicines and School of Pharmacy, Macau University of Science and Technology, Taipa, Macau 999078, China
| | - Xingmei Duan
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Ke Men
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China
| |
Collapse
|
10
|
Masnikosa R, Cvetković Z, Pirić D. Tumor Biology Hides Novel Therapeutic Approaches to Diffuse Large B-Cell Lymphoma: A Narrative Review. Int J Mol Sci 2024; 25:11384. [PMID: 39518937 PMCID: PMC11545713 DOI: 10.3390/ijms252111384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 10/13/2024] [Accepted: 10/16/2024] [Indexed: 11/16/2024] Open
Abstract
Diffuse large B-cell lymphoma (DLBCL) is a malignancy of immense biological and clinical heterogeneity. Based on the transcriptomic or genomic approach, several different classification schemes have evolved over the years to subdivide DLBCL into clinically (prognostically) relevant subsets, but each leaves unclassified samples. Herein, we outline the DLBCL tumor biology behind the actual and potential drug targets and address the challenges and drawbacks coupled with their (potential) use. Therapeutic modalities are discussed, including small-molecule inhibitors, naked antibodies, antibody-drug conjugates, chimeric antigen receptors, bispecific antibodies and T-cell engagers, and immune checkpoint inhibitors. Candidate drugs explored in ongoing clinical trials are coupled with diverse toxicity issues and refractoriness to drugs. According to the literature on DLBCL, the promise for new therapeutic targets lies in epigenetic alterations, B-cell receptor and NF-κB pathways. Herein, we present putative targets hiding in lipid pathways, ferroptosis, and the gut microbiome that could be used in addition to immuno-chemotherapy to improve the general health status of DLBCL patients, thus increasing the chance of being cured. It may be time to devote more effort to exploring DLBCL metabolism to discover novel druggable targets. We also performed a bibliometric and knowledge-map analysis of the literature on DLBCL published from 2014-2023.
Collapse
Affiliation(s)
- Romana Masnikosa
- Department of Physical Chemistry, Vinca Institute of Nuclear Sciences—National Institute of the Republic of Serbia, University of Belgrade, Mike Petrovica Alasa 12-14, 11000 Belgrade, Serbia;
| | - Zorica Cvetković
- Department of Hematology, Clinical Hospital Centre Zemun, Vukova 9, 11000 Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, Dr Subotića 8, 11000 Belgrade, Serbia
| | - David Pirić
- Department of Physical Chemistry, Vinca Institute of Nuclear Sciences—National Institute of the Republic of Serbia, University of Belgrade, Mike Petrovica Alasa 12-14, 11000 Belgrade, Serbia;
| |
Collapse
|
11
|
Cao Y, Bi L, Chen Q, Liu Y, Zhao H, Jin L, Peng R. Understanding the links between micro/nanoplastics-induced gut microbes dysbiosis and potential diseases in fish: A review. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 352:124103. [PMID: 38734053 DOI: 10.1016/j.envpol.2024.124103] [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: 03/16/2024] [Revised: 04/29/2024] [Accepted: 05/02/2024] [Indexed: 05/13/2024]
Abstract
At present, the quantity of micro/nano plastics in the environment is steadily rising, and their pollution has emerged as a global environmental issue. The tendency of their bioaccumulation in aquatic organisms (especially fish) has intensified people's attention to their persistent ecotoxicology. This review critically studies the accumulation of fish in the intestines of fish through active or passive intake of micro/nano plastics, resulting in their accumulation in intestinal organs and subsequent disturbance of intestinal microflora. The key lies in the complex toxic effect on the host after the disturbance of fish intestinal microflora. In addition, this review pointed out the characteristics of micro/nano plastics and the effects of their combined toxicity with adsorbed pollutants on fish intestinal microorganisms, in order to fully understand the characteristics of micro/nano plastics and emphasize the complex interaction between MNPs and other pollutants. We have an in-depth understanding of MNPs-induced intestinal flora disorders and intestinal dysfunction, affecting the host's systemic system, including immune system, nervous system, and reproductive system. The review also underscores the imperative for future research to investigate the toxic effects of prolonged exposure to MNPs, which are crucial for evaluating the ecological risks posed by MNPs and devising strategies to safeguard aquatic organisms.
Collapse
Affiliation(s)
- Yu Cao
- Institute of Life Sciences & Biomedicine Collaborative Innovation Center of Zhejiang province, College of Life and Environmental Science, Wenzhou University, Wenzhou, 325035, China
| | - Liuliu Bi
- Institute of Life Sciences & Biomedicine Collaborative Innovation Center of Zhejiang province, College of Life and Environmental Science, Wenzhou University, Wenzhou, 325035, China
| | - Qianqian Chen
- Institute of Life Sciences & Biomedicine Collaborative Innovation Center of Zhejiang province, College of Life and Environmental Science, Wenzhou University, Wenzhou, 325035, China
| | - Yinai Liu
- Institute of Life Sciences & Biomedicine Collaborative Innovation Center of Zhejiang province, College of Life and Environmental Science, Wenzhou University, Wenzhou, 325035, China
| | - Haiyang Zhao
- Institute of Life Sciences & Biomedicine Collaborative Innovation Center of Zhejiang province, College of Life and Environmental Science, Wenzhou University, Wenzhou, 325035, China
| | - Libo Jin
- Institute of Life Sciences & Biomedicine Collaborative Innovation Center of Zhejiang province, College of Life and Environmental Science, Wenzhou University, Wenzhou, 325035, China
| | - Renyi Peng
- Institute of Life Sciences & Biomedicine Collaborative Innovation Center of Zhejiang province, College of Life and Environmental Science, Wenzhou University, Wenzhou, 325035, China.
| |
Collapse
|
12
|
Huang Z, Huang X, Huang Y, Liang K, Chen L, Zhong C, Chen Y, Chen C, Wang Z, He F, Qin M, Long C, Tang B, Huang Y, Wu Y, Mo X, Weizhong T, Liu J. Identification of KRAS mutation-associated gut microbiota in colorectal cancer and construction of predictive machine learning model. Microbiol Spectr 2024; 12:e0272023. [PMID: 38572984 PMCID: PMC11064510 DOI: 10.1128/spectrum.02720-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 02/27/2024] [Indexed: 04/05/2024] Open
Abstract
Gut microbiota has demonstrated an increasingly important role in the onset and development of colorectal cancer (CRC). Nonetheless, the association between gut microbiota and KRAS mutation in CRC remains enigmatic. We conducted 16S rRNA sequencing on stool samples from 94 CRC patients and employed the linear discriminant analysis effect size algorithm to identify distinct gut microbiota between KRAS mutant and KRAS wild-type CRC patients. Transcriptome sequencing data from nine CRC patients were transformed into a matrix of immune infiltrating cells, which was then utilized to explore KRAS mutation-associated biological functions, including Gene Ontology items and Kyoto Encyclopedia of Genes and Genomes pathways. Subsequently, we analyzed the correlations among these KRAS mutation-associated gut microbiota, host immunity, and KRAS mutation-associated biological functions. At last, we developed a predictive random forest (RF) machine learning model to predict the KRAS mutation status in CRC patients, based on the gut microbiota associated with KRAS mutation. We identified a total of 26 differential gut microbiota between both groups. Intriguingly, a significant positive correlation was observed between Bifidobacterium spp. and mast cells, as well as between Bifidobacterium longum and chemokine receptor CX3CR1. Additionally, we also observed a notable negative correlation between Bifidobacterium and GOMF:proteasome binding. The RF model constructed using the KRAS mutation-associated gut microbiota demonstrated qualified efficacy in predicting the KRAS phenotype in CRC. Our study ascertained the presence of 26 KRAS mutation-associated gut microbiota in CRC and speculated that Bifidobacterium may exert an essential role in preventing CRC progression, which appeared to correlate with the upregulation of mast cells and CX3CR1 expression, as well as the downregulation of GOMF:proteasome binding. Furthermore, the RF model constructed on the basis of KRAS mutation-associated gut microbiota exhibited substantial potential in predicting KRAS mutation status in CRC patients.IMPORTANCEGut microbiota has emerged as an essential player in the onset and development of colorectal cancer (CRC). However, the relationship between gut microbiota and KRAS mutation in CRC remains elusive. Our study not only identified a total of 26 gut microbiota associated with KRAS mutation in CRC but also unveiled their significant correlations with tumor-infiltrating immune cells, immune-related genes, and biological pathways (Gene Ontology items and Kyoto Encyclopedia of Genes and Genomes pathways). We speculated that Bifidobacterium may play a crucial role in impeding CRC progression, potentially linked to the upregulation of mast cells and CX3CR1 expression, as well as the downregulation of GOMF:Proteasome binding. Furthermore, based on the KRAS mutation-associated gut microbiota, the RF model exhibited promising potential in the prediction of KRAS mutation status for CRC patients. Overall, the findings of our study offered fresh insights into microbiological research and clinical prediction of KRAS mutation status for CRC patients.
Collapse
Affiliation(s)
- Zigui Huang
- Division of Colorectal & Anal Surgery, Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Xiaoliang Huang
- Division of Colorectal & Anal Surgery, Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Yili Huang
- College of Oncology, Guangxi Medical University, Nanning, China
| | - Kunmei Liang
- College of Oncology, Guangxi Medical University, Nanning, China
| | - Lei Chen
- College of Oncology, Guangxi Medical University, Nanning, China
| | - Chuzhuo Zhong
- College of Oncology, Guangxi Medical University, Nanning, China
| | - Yingxin Chen
- College of Oncology, Guangxi Medical University, Nanning, China
| | - Chuanbin Chen
- Division of Colorectal & Anal Surgery, Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Zhen Wang
- Division of Colorectal & Anal Surgery, Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Fuhai He
- Division of Colorectal & Anal Surgery, Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Mingjian Qin
- Division of Colorectal & Anal Surgery, Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Chenyan Long
- Division of Colorectal & Anal Surgery, Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Binzhe Tang
- Division of Colorectal & Anal Surgery, Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Yongqi Huang
- Division of Colorectal & Anal Surgery, Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Yongzhi Wu
- Division of Colorectal & Anal Surgery, Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Xianwei Mo
- Division of Colorectal & Anal Surgery, Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Tang Weizhong
- Division of Colorectal & Anal Surgery, Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Jungang Liu
- Division of Colorectal & Anal Surgery, Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| |
Collapse
|
13
|
He J, Li X, Yan M, Chen X, Sun C, Tan J, Song Y, Xu H, Wu L, Yang Z. Inulin Reduces Kidney Damage in Type 2 Diabetic Mice by Decreasing Inflammation and Serum Metabolomics. J Diabetes Res 2024; 2024:1222395. [PMID: 38725443 PMCID: PMC11081752 DOI: 10.1155/2024/1222395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 03/28/2024] [Accepted: 03/30/2024] [Indexed: 05/12/2024] Open
Abstract
This study is aimed at assessing the impact of soluble dietary fiber inulin on the treatment of diabetes-related chronic inflammation and kidney injury in mice with type 2 diabetes (T2DM). The T2DM model was created by feeding the Institute of Cancer Research (ICR) mice a high-fat diet and intraperitoneally injecting them with streptozotocin (50 mg/kg for 5 consecutive days). The thirty-six ICR mice were divided into three dietary groups: the normal control (NC) group, the T2DM (DM) group, and the DM + inulin diet (INU) group. The INU group mice were given inulin at the dose of 500 mg/kg gavage daily until the end of the 12th week. After 12 weeks, the administration of inulin resulted in decreased serum levels of fasting blood glucose (FBG), low-density lipoprotein cholesterol (LDL-C), blood urea nitrogen (BUN), and creatinine (CRE). The administration of inulin not only ameliorated renal injury but also resulted in a reduction in the mRNA expressions of inflammatory factors in the spleen and serum oxidative stress levels, when compared to the DM group. Additionally, inulin treatment in mice with a T2DM model led to a significant increase in the concentrations of three primary short-chain fatty acids (SCFAs) (acetic acid, propionic acid, and butyric acid), while the concentration of advanced glycation end products (AGEs), a prominent inflammatory factor in diabetes, exhibited a significant decrease. The results of untargeted metabolomics indicate that inulin has the potential to alleviate inflammatory response and kidney damage in diabetic mice. This beneficial effect is attributed to its impact on various metabolic pathways, including glycerophospholipid metabolism, taurine and hypotaurine metabolism, arginine biosynthesis, and tryptophan metabolism. Consequently, oral inulin emerges as a promising treatment option for diabetes and kidney injury.
Collapse
Affiliation(s)
- Jiayuan He
- Health Testing Center, Zhenjiang Center for Disease Control and Prevention, Zhenjiang 212002, China
| | - Xiang Li
- Medical Laboratory Department, Huai'an Second People's Hospital, Huai'an 223022, China
| | - Man Yan
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Xinsheng Chen
- Hospital Infection-Disease Control Department, Zhenjiang First People's Hospital, Zhenjiang 212002, China
| | - Chang Sun
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Jiajun Tan
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Yinsheng Song
- Health Testing Center, Zhenjiang Center for Disease Control and Prevention, Zhenjiang 212002, China
| | - Hong Xu
- Health Testing Center, Zhenjiang Center for Disease Control and Prevention, Zhenjiang 212002, China
| | - Liang Wu
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Zhengnan Yang
- Department of Clinical Laboratory, Yizheng Hospital, Nanjing Drum Tower Hospital Group, Yizheng 210008, China
| |
Collapse
|
14
|
Feng Z, Ye W, Feng L. Bioactives and metabolites of Tetrastigma hemsleyanum root extract alleviate DSS-induced ulcerative colitis by targeting the SYK protein in the B cell receptor signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2024; 322:117563. [PMID: 38104876 DOI: 10.1016/j.jep.2023.117563] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 11/27/2023] [Accepted: 12/05/2023] [Indexed: 12/19/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Tetrastigma hemsleyanum is an endemic Chinese herb with a wide range of pharmacological activities, including anti-inflammatory, antiviral, antioxidant, antitumor, and immunomodulatory activities. However, the effect and mechanisms of the anti-inflammatory activity of T. hemsleyanum root extract against dextran sodium sulfate (DSS)-induced ulcerative colitis (UC) have not yet been fully investigated. AIM OF THE STUDY This study aimed to explore the therapeutic effect and molecular mechanisms of T. hemsleyanum root extract in DSS-induced UC mice and knockdown cells. MATERIALS AND METHODS T. hemsleyanum root extract was obtained and analyzed by high-performance liquid chromatography (HPLC). The therapeutic effects of T. hemsleyanum root extract on DSS-induced UC mice were evaluated by the disease activity index (DAI) score, colon length, serum inflammatory cytokines and oxidant/antioxidant levels, and histopathological features of the ileum and colon. Genome-wide gene expression profiles of ileal and colonic tissues were collected by transcriptomics, and signaling pathways were analyzed by the KEGG database. UC-related pathways were uploaded to the STRING database, then the protein-protein interactions (PPIs) were determined by Cytoscape, and the enriched genes were evaluated by real-time quantitative PCR (qPCR). The protein-ligand complexes were docked by AutoDock, and the genes were knocked down in Caco-2 cells by shRNA. The non-targeted metabolomic profiling of ileal contents was analyzed by ultra-high-performance liquid chromatography (UHPLC), and gut microflora were sequenced by an Illumina MiSeq System. RESULTS Ten components that alleviated UC symptoms in mice by decreasing the DAI and serum inflammatory cytokines and oxidant levels, promoting intestinal development, and increasing serum antioxidant levels were identified in T. hemsleyanum root extract. T. hemsleyanum root extract activated the B cell receptor signaling pathway in the colon tissue of UC mice, in which two components, rutin and astragaline, bound to the spleen tyrosine kinase (SYK) protein but also restored gut microflora diversity and increased the proportion of probiotics. Furthermore, metabolites of T. hemsleyanum root extract were involved in vitamin metabolism, fatty acid metabolism, and ferroptosis. CONCLUSIONS The rutin and astragaline components of T. hemsleyanum root extract, by binding to SYK protein, activated the B cell receptor signaling pathway and restored gut microflora diversity to alleviate UC symptoms in mice.
Collapse
Affiliation(s)
- Zhengquan Feng
- Department of Oncology, Tongde Hospital of Zhejiang Province, Hangzhou, 310012, China
| | - Wei Ye
- Food Safety Key Laboratory of Zhejiang Province, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, 310018, China
| | - Lifang Feng
- Food Safety Key Laboratory of Zhejiang Province, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, 310018, China.
| |
Collapse
|
15
|
Fan C, Xu J, Tong H, Fang Y, Chen Y, Lin Y, Chen R, Chen F, Wu G. Gut-brain communication mediates the impact of dietary lipids on cognitive capacity. Food Funct 2024; 15:1803-1824. [PMID: 38314832 DOI: 10.1039/d3fo05288e] [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: 02/07/2024]
Abstract
Cognitive impairment, as a prevalent symptom of nervous system disorders, poses one of the most challenging aspects in the management of brain diseases. Lipids present in the cell membranes of all neurons within the brain and dietary lipids can regulate the cognition and memory function. In recent years, the advancements in gut microbiome research have enabled the exploration of dietary lipids targeting the gut-brain axis as a strategy for regulating cognition. This present review provides an in-depth overview of how lipids modulate cognition via the gut-brain axis depending on metabolic, immune, neural and endocrine pathways. It also comprehensively analyzes the effects of diverse lipids on the gut microbiota and intestinal barrier function, thereby affecting the central nervous system and cognitive capacity. Moreover, comparative analysis of the positive and negative effects is presented between beneficial and detrimental lipids. The former encompass monounsaturated fatty acids, short-chain fatty acids, omega-3 polyunsaturated fatty acids, phospholipids, phytosterols, fungal sterols and bioactive lipid-soluble vitamins, as well as lipid-derived gut metabolites, whereas the latter (detrimental lipids) include medium- or long-chain fatty acids, excessive proportions of n-6 polyunsaturated fatty acids, industrial trans fatty acids, and zoosterols. To sum up, the focus of this review is on how gut-brain communication mediates the impact of dietary lipids on cognitive capacity, providing a novel theoretical foundation for promoting brain cognitive health and scientific lipid consumption patterns.
Collapse
Affiliation(s)
- Chenhan Fan
- School of Public Health, Health Science Center, Ningbo University, Ningbo, Zhejiang, 315211, China.
| | - Jingxuan Xu
- School of Public Health, Health Science Center, Ningbo University, Ningbo, Zhejiang, 315211, China.
| | - Haoxiang Tong
- School of Public Health, Health Science Center, Ningbo University, Ningbo, Zhejiang, 315211, China.
| | - Yucheng Fang
- School of Public Health, Health Science Center, Ningbo University, Ningbo, Zhejiang, 315211, China.
| | - Yiming Chen
- School of Public Health, Health Science Center, Ningbo University, Ningbo, Zhejiang, 315211, China.
| | - Yangzhuo Lin
- School of Basic Medical Science, Health Science Center, Ningbo University, Ningbo, Zhejiang, 315211, P. R. China
| | - Rui Chen
- School of Basic Medical Science, Health Science Center, Ningbo University, Ningbo, Zhejiang, 315211, P. R. China
| | - Fuhao Chen
- School of Basic Medical Science, Health Science Center, Ningbo University, Ningbo, Zhejiang, 315211, P. R. China
| | - Guoqing Wu
- School of Public Health, Health Science Center, Ningbo University, Ningbo, Zhejiang, 315211, China.
| |
Collapse
|
16
|
Wen X, Yang H, Li Z, Chu W. Alcohol degradation, learning, and memory-enhancing effect of Acetobacter pasteurianus BP2201 in Caenorhabditis elegans model. J Appl Microbiol 2023; 134:lxad253. [PMID: 37934610 DOI: 10.1093/jambio/lxad253] [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: 07/23/2023] [Revised: 08/18/2023] [Accepted: 10/31/2023] [Indexed: 11/09/2023]
Abstract
AIMS This study aimed to investigate the probiotic effects of Acetobacter pasteurianus BP2201, isolated from brewing mass, for the treatment of alcohol-induced learning and memory ability impairments in a Caenorhabditis elegans model. METHODS AND RESULTS Acetobacter pasteurianus BP2201 was examined for probiotic properties, including acid and bile salt resistance, ethanol degradation, antioxidant efficacy, hemolytic activity, and susceptibility to antibiotics. The strain displayed robust acid and bile salt tolerance, efficient ethanol degradation, potent antioxidant activity, and susceptibility to specific antibiotics. Additionally, in the C. elegans model, administering A. pasteurianus BP2201 significantly improved alcohol-induced learning and memory impairments. CONCLUSIONS Acetobacter pasteurianus BP2201 proves to be a promising candidate strain for the treatment of learning and memory impairments induced by alcohol intake.
Collapse
Affiliation(s)
- Xin Wen
- Department of Pharmaceutical Microbiology, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
| | - Huazhong Yang
- Department of Pharmaceutical Microbiology, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
| | - Zhongqi Li
- Department of Pharmaceutical Microbiology, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
| | - Weihua Chu
- Department of Pharmaceutical Microbiology, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
17
|
Li P, Chen J, Guo CE, Li W, Gao Z. Lactobacillus co-fermentation of Cerasus humilis juice alters chemical properties, enhances antioxidant activity, and improves gut microbiota. Food Funct 2023; 14:8248-8260. [PMID: 37655677 DOI: 10.1039/d3fo02583g] [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: 09/02/2023]
Abstract
Fermentation with Lactobacillus has been shown to improve the nutritional value of juice. In this study, Cerasus humilis juice was fermented using two commercial probiotics, namely, Lactobacillus acidophilus and Lactobacillus plantarum. The total antioxidant capacity (TAOC), viable count, chemical properties, antioxidant activity after in vitro digestion, and alterations in the gut microbiota composition of the fermented juice were investigated. After fermentation, the TAOC increased from 107.66 U mL-1 to 126.72 U mL-1; viable count increased from 5.85 lg (CFU mL-1) to 8.17 lg (CFU mL-1); and the contents of total phenols, total flavonoids, proanthocyanins, four organic acids, and 29 amino acids had changed. Overall, 47 compounds were identified in the juice, 20 of which were enriched after fermentation. Furthermore, Lactobacillus co-fermentation improved the antioxidant properties of the juice after in vitro digestion and increased the abundance of probiotics to regulate the gut microbiota. These findings illustrate the potential use of Lactobacillus acidophilus and Lactobacillus plantarum in the co-fermentation of C. humilis juice to enhance its nutritional and functional properties.
Collapse
Affiliation(s)
- Ping Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, P. R. China.
| | - Jiaji Chen
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, P. R. China.
| | - Chang-E Guo
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, P. R. China.
| | - Weidong Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, P. R. China.
| | - Zhiliang Gao
- Zhiliang Dute Jingluo Tcm Pte.Ltd., Block509 Bedok North Street 3 460509, Singapore
| |
Collapse
|
18
|
Ben-Azu B, del Re EC, VanderZwaag J, Carrier M, Keshavan M, Khakpour M, Tremblay MÈ. Emerging epigenetic dynamics in gut-microglia brain axis: experimental and clinical implications for accelerated brain aging in schizophrenia. Front Cell Neurosci 2023; 17:1139357. [PMID: 37256150 PMCID: PMC10225712 DOI: 10.3389/fncel.2023.1139357] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 04/27/2023] [Indexed: 06/01/2023] Open
Abstract
Brain aging, which involves a progressive loss of neuronal functions, has been reported to be premature in probands affected by schizophrenia (SCZ). Evidence shows that SCZ and accelerated aging are linked to changes in epigenetic clocks. Recent cross-sectional magnetic resonance imaging analyses have uncovered reduced brain reserves and connectivity in patients with SCZ compared to typically aging individuals. These data may indicate early abnormalities of neuronal function following cyto-architectural alterations in SCZ. The current mechanistic knowledge on brain aging, epigenetic changes, and their neuropsychiatric disease association remains incomplete. With this review, we explore and summarize evidence that the dynamics of gut-resident bacteria can modulate molecular brain function and contribute to age-related neurodegenerative disorders. It is known that environmental factors such as mode of birth, dietary habits, stress, pollution, and infections can modulate the microbiota system to regulate intrinsic neuronal activity and brain reserves through the vagus nerve and enteric nervous system. Microbiota-derived molecules can trigger continuous activation of the microglial sensome, groups of receptors and proteins that permit microglia to remodel the brain neurochemistry based on complex environmental activities. This remodeling causes aberrant brain plasticity as early as fetal developmental stages, and after the onset of first-episode psychosis. In the central nervous system, microglia, the resident immune surveillance cells, are involved in neurogenesis, phagocytosis of synapses and neurological dysfunction. Here, we review recent emerging experimental and clinical evidence regarding the gut-brain microglia axis involvement in SCZ pathology and etiology, the hypothesis of brain reserve and accelerated aging induced by dietary habits, stress, pollution, infections, and other factors. We also include in our review the possibilities and consequences of gut dysbiosis activities on microglial function and dysfunction, together with the effects of antipsychotics on the gut microbiome: therapeutic and adverse effects, role of fecal microbiota transplant and psychobiotics on microglial sensomes, brain reserves and SCZ-derived accelerated aging. We end the review with suggestions that may be applicable to the clinical setting. For example, we propose that psychobiotics might contribute to antipsychotic-induced therapeutic benefits or adverse effects, as well as reduce the aging process through the gut-brain microglia axis. Overall, we hope that this review will help increase the understanding of SCZ pathogenesis as related to chronobiology and the gut microbiome, as well as reveal new concepts that will serve as novel treatment targets for SCZ.
Collapse
Affiliation(s)
- Benneth Ben-Azu
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Department of Pharmacology, Faculty of Basic Medical Sciences, College of Health Sciences, Delta State University, Abraka, Nigeria
| | - Elisabetta C. del Re
- Department of Psychiatry, Harvard Medical School, Boston, MA, United States
- VA Boston Healthcare System, Brockton, MA, United States
- Beth Israel Deaconess Medical Center, Boston, MA, United States
| | - Jared VanderZwaag
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Micaël Carrier
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Matcheri Keshavan
- Department of Psychiatry, Harvard Medical School, Boston, MA, United States
- Beth Israel Deaconess Medical Center, Boston, MA, United States
| | | | - Marie-Ève Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Axe Neurosciences, Centre de Recherche du CHU de Québec, Université Laval, Québec City, QC, Canada
- Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, BC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada
- Department of Molecular Medicine, Université Laval, Québec City, QC, Canada
- Centre for Advanced Materials and Related Technology (CAMTEC), Institute on Aging and Lifelong Health (IALH), University of Victoria, Victoria, BC, Canada
| |
Collapse
|
19
|
Liu W, Xu C, Zou Z, Weng Q, Xiao Y. Sestrin2 suppresses ferroptosis to alleviate septic intestinal inflammation and barrier dysfunction. Immunopharmacol Immunotoxicol 2023; 45:123-132. [PMID: 36066109 DOI: 10.1080/08923973.2022.2121927] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 08/29/2022] [Indexed: 11/12/2022]
Abstract
OBJECTIVE Alterations in intestinal function play a crucial role in the pathogenesis of sepsis, and the repair of the intestinal barrier is a potential strategy for the treatment of sepsis. Sestrin2 (SESN2), a highly conserved stress-responsive protein, can be induced in response to stress. AIM This paper aimed to explore the role and mechanism of SESN2 in septic intestinal dysfunction. METHODS Blood samples were collected from patients with septic intestinal dysfunction, and Caco-2 cells were subjected to lipopolysaccharide (LPS) to construct in vitro models. The expression level of SESN2 was determined in the blood samples and cells. The impacts of SESN2 overexpression on cell inflammation, oxidative stress, barrier integrity, and MAPK/Nrf2 signaling were evaluated. To determine the mediated role of MAPK signaling and ferroptosis, AMPK inhibitor (Compound C) and ferroptosis inducer (erastin) were separately used to treat cells, and the influences on the above aspects in cells were assessed. RESULTS The expression level of SESN2 was down-regulated in patients with septic intestinal dysfunction and LPS-induced cells. SESN2 overexpression was found to suppress cell inflammation and oxidative stress, maintain barrier integrity, and activate AMPK/Nrf2 signaling. Following the AMPK signaling was inhibited or the ferroptosis was triggered, the effects of SESN2 overexpression on the cells were both reversed. CONCLUSION Reduced SESN2 contributed to inflammatory response and barrier dysfunction in septic intestinal dysfunction by promoting ferroptosis via activating the AMPK/Nrf2 signaling pathway.
Collapse
Affiliation(s)
- Wei Liu
- Department of Critical Care Medicine, Fujian Medical University Union Hospital, Fuzhou, P.R. China
| | - Chanchan Xu
- Department of Internal Medicine, Shanghai Raffles Hospital, Shanghai, P.R. China
| | - Zhiqiang Zou
- Department of Critical Care Medicine, Fujian Medical University Union Hospital, Fuzhou, P.R. China
| | - Qinyong Weng
- Department of Critical Care Medicine, Fujian Medical University Union Hospital, Fuzhou, P.R. China
| | - Ying Xiao
- Department of Critical Care Medicine, Fujian Medical University Union Hospital, Fuzhou, P.R. China
| |
Collapse
|
20
|
Zhang H, Zhang H, Jiang Q. Progress in research of gut microbiota in colorectal cancer. Shijie Huaren Xiaohua Zazhi 2023; 31:138-142. [DOI: 10.11569/wcjd.v31.i4.138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/02/2023] Open
Abstract
The human gut microbiota is a large and complex microbial community that is linked to human health and disease. Intestinal homeostasis is dependent on the tight interplay between the host and gut microbiota. Moreover, the gut microbiota plays an important role in digestion and metabolism. In recent years, the gut microbiota is still the most studied topic, and numerous studies have shown that the gut microbiota is closely related to colorectal cancer. In this paper, we will review the relationship between the gut microbiota and colorectal cancer pathogenesis, prevention, and treatment, with an aim to provide some new ideas for the research of colorectal cancer.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Laboratory Medicine, Chengbei District, Hangzhou First People's Hospital, Hangzhou 310022, Zhejiang Province, China
| | - Hong Zhang
- Department of Laboratory Medicine, Chengbei District, Hangzhou First People's Hospital, Hangzhou 310022, Zhejiang Province, China
| | - Qin Jiang
- Department of Laboratory Medicine, Chengbei District, Hangzhou First People's Hospital, Hangzhou 310022, Zhejiang Province, China
| |
Collapse
|
21
|
Liu Q, An X, Chen Y, Deng Y, Niu H, Ma R, Zhao H, Cao W, Wang X, Wang M. Effects of Auricularia auricula Polysaccharides on Gut Microbiota and Metabolic Phenotype in Mice. Foods 2022; 11:foods11172700. [PMID: 36076885 PMCID: PMC9455240 DOI: 10.3390/foods11172700] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/17/2022] [Accepted: 09/01/2022] [Indexed: 11/26/2022] Open
Abstract
Personalized diets change the internal metabolism of organisms, which, in turn, affects the health of the body; this study was performed to explore the regulatory effects of polysaccharides extracted from Auricularia auricula on the overall metabolism and gut microbiota in normal C57BL/6J mice. The study was conducted using metabolomic and microbiomic methods to provide a scientific basis for further development and use of Auricularia auricula resources in the Qinba Mountains and in nutritional food with Auricularia auricula polysaccharides (AAP) as the main functional component. Based on LC-MS/MS metabolomic results, 51 AAP-regulated metabolites were found, mainly enriched in the arginine biosynthesis pathway, which had the highest correlation, followed by the following metabolisms: arginine and proline; glycine, serine and threonine; and glycerophospholipid, along with the sphingolipid metabolism pathway. Furthermore, supplementation of AAP significantly changed the composition of the mice intestinal flora. The relative abundance levels of Lactobacillus johnsonii, Weissella cibaria, Kosakonia cowanii, Enterococcus faecalis, Bifidobacterium animalis and Bacteroides uniformis were markedly up-regulated, while the relative abundance of Firmicutes bacterium M10-2 was down-regulated. The bioactivities of AAP may be related to the regulatory effects of endogenous metabolism and gut microbiota composition.
Collapse
Affiliation(s)
- Qian Liu
- College of Food Science and Technology, Northwest University, 229 Taibai North Road, Xi’an 710069, China
- Shaanxi Functional Food Engineering Center Co., Ltd., Xi’an 710069, China
- Correspondence: ; Tel./Fax: +86-29-88305208
| | - Xin An
- College of Food Science and Technology, Northwest University, 229 Taibai North Road, Xi’an 710069, China
| | - Yuan Chen
- College of Food Science and Technology, Northwest University, 229 Taibai North Road, Xi’an 710069, China
| | - Yuxuan Deng
- College of Food Science and Technology, Northwest University, 229 Taibai North Road, Xi’an 710069, China
| | - Haili Niu
- College of Food Science and Technology, Northwest University, 229 Taibai North Road, Xi’an 710069, China
| | - Ruisen Ma
- College of Food Science and Technology, Northwest University, 229 Taibai North Road, Xi’an 710069, China
| | - Haoan Zhao
- College of Food Science and Technology, Northwest University, 229 Taibai North Road, Xi’an 710069, China
| | - Wei Cao
- College of Food Science and Technology, Northwest University, 229 Taibai North Road, Xi’an 710069, China
| | - Xiaoru Wang
- Shaanxi Functional Food Engineering Center Co., Ltd., Xi’an 710069, China
| | - Meng Wang
- Shaanxi Functional Food Engineering Center Co., Ltd., Xi’an 710069, China
| |
Collapse
|
22
|
Li P, Li M, Song Y, Huang X, Wu T, Xu ZZ, Lu H. Green Banana Flour Contributes to Gut Microbiota Recovery and Improves Colonic Barrier Integrity in Mice Following Antibiotic Perturbation. Front Nutr 2022; 9:832848. [PMID: 35369097 PMCID: PMC8964434 DOI: 10.3389/fnut.2022.832848] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 02/07/2022] [Indexed: 12/12/2022] Open
Abstract
Green banana flour (GBF) is rich in resistant starch that has been used as a prebiotic to exert beneficial effects on gut microbiota. In this study, GBF was evaluated for its capacity to restore gut microbiota and intestinal barrier integrity from antibiotics (Abx) perturbation by comparing it to natural recovery (NR) treatment. C57B/L 6 J mice were exposed to 3 mg ciprofloxacin and 3.5 mg metronidazole once a day for 2 weeks to induce gut microbiota dysbiosis model. Then, GBF intervention at the dose of 400 mg/kg body weight was conducted for 2 weeks. The results showed that mice treated with Abx displayed increased gut permeability and intestinal barrier disruption, which were restored more quickly with GBF than NR treatment by increasing the secretion of mucin. Moreover, GBF treatment enriched beneficial Bacteroidales S24-7, Lachnospiraceae, Bacteroidaceae, and Porphyromonadaceae that accelerated the imbalanced gut microbiota restoration to its original state. This study puts forward novel insights into the application of GBF as a functional food ingredient to repair gut microbiota from Abx perturbation.
Collapse
Affiliation(s)
- Ping Li
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China
| | - Ming Li
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China
| | - Ying Song
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China
| | - Xiaochang Huang
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China
| | - Tao Wu
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China
| | - Zhenjiang Zech Xu
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China
| | - Hui Lu
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China
| |
Collapse
|
23
|
Xiang Q, Cheng L, Zhang R, Liu Y, Wu Z, Zhang X. Tea Polyphenols Prevent and Intervene in COVID-19 through Intestinal Microbiota. Foods 2022; 11:506. [PMID: 35205982 PMCID: PMC8871045 DOI: 10.3390/foods11040506] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/22/2022] [Accepted: 02/05/2022] [Indexed: 12/13/2022] Open
Abstract
Although all countries have taken corresponding measures, the coronavirus disease 2019 (COVID-19) is still ravaging the world. To consolidate the existing anti-epidemic results and further strengthen the prevention and control measures against the new coronavirus, we are now actively pioneering a novel research idea of regulating the intestinal microbiota through tea polyphenols for reference. Although studies have long revealed the regulatory effect of tea polyphenols on the intestinal microbiota to various gastrointestinal inflammations, little is known about the prevention and intervention of COVID-19. This review summarizes the possible mechanism of the influence of tea polyphenols on COVID-19 mediated by the intestinal microbiota. In this review, the latest studies of tea polyphenols exhibiting their own antibacterial and anti-inflammatory activities and protective effects on the intestinal mucosal barrier are combed through and summarized. Among them, (-)-epigallocatechin-3-gallate (EGCG), one of the main monomers of catechins, may be activated as nuclear factor erythroid 2 p45-related factor 2 (Nrf2). The agent inhibits the expression of ACE2 (a cellular receptor for SARS-CoV-2) and TMPRSS2 to inhibit SARS-CoV-2 infection, inhibiting the life cycle of SARS-CoV-2. Thus, preliminary reasoning and judgments have been made about the possible mechanism of the effect of tea polyphenols on the COVID-19 control and prevention mediated by the microbiota. These results may be of great significance to the future exploration of specialized research in this field.
Collapse
Affiliation(s)
- Qiao Xiang
- Department of Food Science and Engineering, Ningbo University, Ningbo 315211, China; (Q.X.); (Y.L.); (Z.W.)
| | - Lu Cheng
- Department of Food Science, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA;
| | - Ruilin Zhang
- Key Laboratory of Animal Protein Deep Processing Technology of Zhejiang Province, Ningbo University, Ningbo 315211, China
| | - Yanan Liu
- Department of Food Science and Engineering, Ningbo University, Ningbo 315211, China; (Q.X.); (Y.L.); (Z.W.)
| | - Zufang Wu
- Department of Food Science and Engineering, Ningbo University, Ningbo 315211, China; (Q.X.); (Y.L.); (Z.W.)
| | - Xin Zhang
- Department of Food Science and Engineering, Ningbo University, Ningbo 315211, China; (Q.X.); (Y.L.); (Z.W.)
| |
Collapse
|