1
|
Xu Q, Li Y, Qin X, Xin Y, Wang J, Zhang Y, Xu K, Yang X, Wang X. osa-miR168a, a Plant miRNA That Survives the Process of In Vivo Food Digestion, Attenuates Dextran Sulfate Sodium-Induced Colitis in Mice by Oral Administration. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024. [PMID: 39480689 DOI: 10.1021/acs.jafc.4c07283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2024]
Abstract
Previous studies showed that osa-miR168a, a plant miRNA rich in fruits and vegetables, had cross-kingdom biological effects on immunocytes, silkworms, and rodents. In this study, the effects of miR168a on mouse colitis induced by dextran sulfate sodium (DSS) were investigated. The results showed that miR168a oligomers were resistant during the process of food digestion, ending up with a residual concentration of 67.8 ± 11.2 fM in mouse intestines 4 h after oral gavage. More importantly, direct oral administration of the miRNA to the colitis mice significantly ameliorated the progression of the disease, as evidenced by the reduction in DAI score, histopathological lesions, and proinflammatory cytokines. Repairing intestinal barrier function by promoting the regeneration of TJ proteins and the mucus layer, suppressing oxidative stress and colonic inflammation via modulating Nrf2 and NF-κB signaling pathways, and restoring the imbalanced gut microbiota caused by DSS are proposed mechanisms behind the anticolitis activity of miR168a. This study provided new evidence of the cross-kingdom regulatory effects of dietary miRNAs, suggesting the potential of the plant miRNA for the prevention and treatment of inflammatory bowel diseases.
Collapse
Affiliation(s)
- Qin Xu
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, Shaanxi 710062, China
| | - Ying Li
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, Shaanxi 710062, China
| | - Xinshu Qin
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, Shaanxi 710062, China
- Department of Nutrition and Health, China Agricultural University, Beijing 100193, China
| | - Yirao Xin
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, Shaanxi 710062, China
| | - Jianing Wang
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, Shaanxi 710062, China
| | - Yi Zhang
- Department of Food Science, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Ke Xu
- Department of Joint Surgery, Hong Hui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710054, China
| | - Xingbin Yang
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, Shaanxi 710062, China
| | - Xingyu Wang
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, Shaanxi 710062, China
| |
Collapse
|
2
|
Zheng C, Song J, Shan M, Qiu M, Cui M, Huang C, Chen W, Wang J, Zhang L, Yu Y, Fang H. Key bacterial taxa with specific metabolisms and life history strategies sustain soil microbial network stability exposed to carbendazim and deoxynivalenol. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 954:176680. [PMID: 39366579 DOI: 10.1016/j.scitotenv.2024.176680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 09/23/2024] [Accepted: 09/30/2024] [Indexed: 10/06/2024]
Abstract
Co-contamination of carbendazim (CBD) and deoxynivalenol (DON) is common in agricultural soils, yet their ecological impact on soil microbiome remains poorly assessed. Here, we investigated the influence of CBD and DON on the structure, function, and co-occurrence networks of soil microbiome. The combined treatment of CBD and DON significantly exacerbated the negative impacts on soil microbial diversity, functional diversity, and microbial network stability compared to individual treatments. Specifically, Lysobacter, Gemmatimonas, Nitrospira, Massilia, and Bacillus were identified as indicator species for CBD and DON. Simultaneously, the abundance of genes involved in key ecological functions, such as nitrification (amoA) and organic phosphorus mineralization (phoAD), was significantly reduced. Notably, key bacterial taxa Nitrospira and Gemmatimonas, with K-life history strategy and capabilities for nitrification and organic nitrogen mineralization, played crucial roles in promoting positive interactions in networks. Furthermore, variance partitioning analysis (VPA) and structural equation modeling (SEM) demonstrated that the abundance and niche breadth of key bacterial taxa were the primary drivers of microbial network stability. In conclusion, our study provides new insights into how soil microbiomes and networks respond to pesticides and mycotoxins, aiding in a more comprehensive assessment of exposure risks.
Collapse
Affiliation(s)
- Conglai Zheng
- Zhejiang Key Laboratory of Biology and Ecological Regulation of Crop Pathogens and Insects, Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Ministry of Agriculture and Rural Affairs, Institute of Pesticide and Environmental Toxicology, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou 310058, China
| | - Jiajin Song
- Zhejiang Key Laboratory of Biology and Ecological Regulation of Crop Pathogens and Insects, Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Ministry of Agriculture and Rural Affairs, Institute of Pesticide and Environmental Toxicology, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou 310058, China
| | - Mei Shan
- Zhejiang Key Laboratory of Biology and Ecological Regulation of Crop Pathogens and Insects, Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Ministry of Agriculture and Rural Affairs, Institute of Pesticide and Environmental Toxicology, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou 310058, China
| | - Mengting Qiu
- Zhejiang Key Laboratory of Biology and Ecological Regulation of Crop Pathogens and Insects, Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Ministry of Agriculture and Rural Affairs, Institute of Pesticide and Environmental Toxicology, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou 310058, China
| | - Minrong Cui
- Zhejiang Key Laboratory of Biology and Ecological Regulation of Crop Pathogens and Insects, Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Ministry of Agriculture and Rural Affairs, Institute of Pesticide and Environmental Toxicology, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou 310058, China
| | - Chenyu Huang
- Zhejiang Key Laboratory of Biology and Ecological Regulation of Crop Pathogens and Insects, Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Ministry of Agriculture and Rural Affairs, Institute of Pesticide and Environmental Toxicology, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou 310058, China
| | - Weibin Chen
- Zhejiang Key Laboratory of Biology and Ecological Regulation of Crop Pathogens and Insects, Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Ministry of Agriculture and Rural Affairs, Institute of Pesticide and Environmental Toxicology, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou 310058, China
| | - Jiao Wang
- Zhejiang Key Laboratory of Biology and Ecological Regulation of Crop Pathogens and Insects, Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Ministry of Agriculture and Rural Affairs, Institute of Pesticide and Environmental Toxicology, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou 310058, China
| | - Luqing Zhang
- Zhejiang Key Laboratory of Biology and Ecological Regulation of Crop Pathogens and Insects, Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Ministry of Agriculture and Rural Affairs, Institute of Pesticide and Environmental Toxicology, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou 310058, China
| | - Yunlong Yu
- Zhejiang Key Laboratory of Biology and Ecological Regulation of Crop Pathogens and Insects, Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Ministry of Agriculture and Rural Affairs, Institute of Pesticide and Environmental Toxicology, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou 310058, China
| | - Hua Fang
- Zhejiang Key Laboratory of Biology and Ecological Regulation of Crop Pathogens and Insects, Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Ministry of Agriculture and Rural Affairs, Institute of Pesticide and Environmental Toxicology, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
3
|
Zhao Y, Guo K, Yan Y, Jiang B. Cucurbitacin IIb alleviates colitis via regulating gut microbial composition and metabolites. Heliyon 2024; 10:e38051. [PMID: 39347394 PMCID: PMC11437856 DOI: 10.1016/j.heliyon.2024.e38051] [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: 03/03/2024] [Revised: 09/03/2024] [Accepted: 09/17/2024] [Indexed: 10/01/2024] Open
Abstract
Cucurbitacin IIb, a member of the triterpenoid family, exerts beneficial effects on intestinal diseases, including enteritis and bacillary dysentery. However, its effects and mechanisms of action on colitis have not yet been explored. In this study, we used a mouse model of dextran sulfate sodium (DSS)-induced colitis and explored the effects of cucurbitacin IIb on colitis symptoms, inflammatory responses, microbiota, and metabolite profiles. The results showed that cucurbitacin IIb alleviated colitis symptoms including body weight loss, an increase in the disease activity index, and elevated levels of myeloperoxidase and eosinophil peroxidase content. Additionally, it ameliorated intestinal morphology impairment, reduced the phosphorylation of NFκB protein, and mitigated accumulation of pro-inflammatory cytokines IL-6 and IL-1β. Furthermore, cucurbitacin IIb alleviated alterations in gut microbial composition and metabolites in DSS-treated mice. However, antibiotic treatment diminishes the beneficial effects of cucurbitacin IIb on colitis. We further found that transplantation of fresh feces or heat-inactivated feces from mice treated with cucurbitacin IIb to DSS-treated mice alleviated colitis, similar to the effects of cucurbitacin IIb. Collectively, our results suggest that cucurbitacin IIb exerted anti-inflammatory effects in colitis by regulating the microbiota composition and metabolites, thereby alleviating colitis symptoms.
Collapse
Affiliation(s)
- Yinyin Zhao
- Ningbo Institute of Innovation for Combined Medicine and Engineering (NIIME), The Affiliated LiHuiLi Hospital of Ningbo University, Ningbo, 315000, China
| | - Kangxiao Guo
- Pharmaceutical College, Changsha Health Vocational College, Changsha, 410699, China
| | - Yongwang Yan
- Pharmaceutical College, Changsha Health Vocational College, Changsha, 410699, China
| | - Binyuan Jiang
- Medical Research Center, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, 410004, China
| |
Collapse
|
4
|
Zhang Y, Zhao M, Li Y, Liang S, Li X, Wu Y, Li G. Potential Probiotic Properties and Complete Genome Analysis of Limosilactobacillus reuteri LRA7 from Dogs. Microorganisms 2024; 12:1811. [PMID: 39338485 DOI: 10.3390/microorganisms12091811] [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: 08/12/2024] [Revised: 08/23/2024] [Accepted: 08/29/2024] [Indexed: 09/30/2024] Open
Abstract
This study aimed to isolate and screen canine-derived probiotics with excellent probiotic properties. Strain characterization was conducted using a combination of in vitro and in vivo probiotic characterization and safety assessments, as well as complete genome analysis. The results showed that Limosilactobacillus reuteri LRA7 exhibited excellent bacteriostatic and antioxidant activities. The survival rate at pH 2.5 was 79.98%, and the viable counts after exposure to gastrointestinal fluid and 0.5% bile salts were 7.77 log CFU/mL and 5.29 log CFU/mL, respectively. The bacterium also exhibited high hydrophobicity, self-coagulation, and high temperature tolerance, was negative for hemolysis, and was sensitive to clindamycin. In vivo studies in mice showed that the serum superoxide dismutase activity level was 53.69 U/mL higher in the MR group of mice compared to that of the control group, the malondialdehyde content was 0.53 nmol/mL lower in the HR group, and the highest jejunal V/C value was 4.11 ± 1.05 in the HR group (p < 0.05). The L. reuteri LRA7 gene is 2.021 megabases in size, contains one chromosome and one plasmid, and is annotated with 1978 functional genes. In conclusion, L. reuteri LRA7 has good probiotic potential and is safe. It can be used as an ideal probiotic candidate strain of canine origin.
Collapse
Affiliation(s)
- Yuanyuan Zhang
- College of Animal Science and Technology, Qingdao Agricultural University, Qingdao 266109, China
| | - Mengdi Zhao
- College of Animal Science and Technology, Qingdao Agricultural University, Qingdao 266109, China
- College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, China
| | - Yueyao Li
- College of Animal Science and Technology, Qingdao Agricultural University, Qingdao 266109, China
| | - Shuang Liang
- College of Animal Science and Technology, Qingdao Agricultural University, Qingdao 266109, China
| | - Xinkang Li
- College of Animal Science and Technology, Qingdao Agricultural University, Qingdao 266109, China
| | - Yi Wu
- College of Animal Science and Technology, Qingdao Agricultural University, Qingdao 266109, China
| | - Guangyu Li
- College of Animal Science and Technology, Qingdao Agricultural University, Qingdao 266109, China
| |
Collapse
|
5
|
Liang J, Zeng Y, Hu H, Yin Y, Zhou X. Prevotella copri Improves Selenium Deposition and Meat Quality in the longissimus dorsi Muscle of Fattening Pigs. Probiotics Antimicrob Proteins 2024:10.1007/s12602-024-10340-1. [PMID: 39105886 DOI: 10.1007/s12602-024-10340-1] [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] [Accepted: 07/27/2024] [Indexed: 08/07/2024]
Abstract
Selenium is among the important trace elements that influence the quality of meat. Although it has been established that the gut microbiota is closely associated with selenium metabolism, it has yet to be determined whether these microbes influence the accumulation of selenium in muscles. To identify gut microbiota that potentially influence the deposition of selenium in muscles, we compared the colonic microbial composition of pigs characterized by high and low contents of selenium in the longissimus dorsi muscle and accordingly detected a higher abundance of the bacterium Prevotella copri (P. copri) in pigs with a higher muscle selenium content. To verify the effect of P. copri, 16 pigs weighing approximately 61 kg were fed either a basal diet or a basal diet supplemented with P. copri (1.0 × 1010 CFU/kg feed) for 45 days. The results revealed significant increases in the contents of selenium and selenoprotein in the serum and longissimus dorsi muscle of fattening pigs fed the P. copri-supplemented diet. Moreover, supplementing the feed of pigs with P. copri was observed to promote significant improvement in the antioxidant capacity and quality of meat, including drip loss, pH, and meat color. In conclusion, our findings in this study indicate that P. copri has potential utility as a dietary supplement for improving the selenium status and meat quality in fattening pigs.
Collapse
Affiliation(s)
- Jing Liang
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture the Chinese Academy of Sciences, Changsha, 410125, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, 101408, China
| | - Yan Zeng
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture the Chinese Academy of Sciences, Changsha, 410125, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, 101408, China
| | - Hong Hu
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, 410128, China
| | - Yulong Yin
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture the Chinese Academy of Sciences, Changsha, 410125, China.
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, 101408, China.
| | - Xihong Zhou
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture the Chinese Academy of Sciences, Changsha, 410125, China.
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, 101408, China.
- Hunan Provincial Key Laboratory of the Traditional Chinese Medicine Agricultural Biogenomics, Changsha Medical University, Changsha, 410208, China.
| |
Collapse
|
6
|
Zha A, Li W, Wang J, Bai P, Qi M, Liao P, Tan B, Yin Y. Trimethylamine oxide supplementation differentially regulates fat deposition in liver, longissimus dorsi muscle and adipose tissue of growing-finishing pigs. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2024; 17:25-35. [PMID: 38464952 PMCID: PMC10920132 DOI: 10.1016/j.aninu.2023.12.006] [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: 03/08/2023] [Revised: 12/22/2023] [Accepted: 12/29/2023] [Indexed: 03/12/2024]
Abstract
Trimethylamine oxide (TMAO) is a microbiota-derived metabolite, and numerous studies have shown that it could regulate fat metabolism in humans and mice. However, few studies have focused on the effects of TMAO on fat deposition in growing-finishing pigs. This study aimed to investigate the effect of TMAO on fat deposition and intestinal microbiota in growing-finishing pigs. Sixteen growing pigs were randomly divided into 2 groups and fed with a basal diet with 0 or 1 g/kg TMAO for 149 d. The intestinal microbial profiles, fat deposition indexes, and fatty acid profiles were measured. These results showed that TMAO supplementation had a tendency to decrease lean body mass (P < 0.1) and significantly increased backfat thickness (P < 0.05), but it did not affect growth performance. TMAO significantly increased total protein (TP) concentration, and reduced alkaline phosphatase (ALP) concentration in serum (P < 0.05). TMAO increased the α diversity of the ileal microbiota community (P < 0.05), and it did not affect the colonic microbial community. TMAO supplementation significantly increased acetate content in the ileum, and Proteobacteria and Escherichia-Shigella were significantly enriched in the TMAO group (P < 0.05). In addition, TMAO decreased fat content, as well as the ratio of linoleic acid, n-6 polyunsaturated fatty acids (PUFA), and PUFA in the liver (P < 0.05). On the contrary, TMAO increased intramuscular fat content of the longissimus dorsi muscle, whereas the C18:2n6c ratio was increased, and the n-6 PUFA:PUFA ratio was decreased (P < 0.05). In vitro, 1 mM TMAO treatment significantly upregulated the expression of FASN and SREBP1 in C2C12 cells (P < 0.05). Nevertheless, TMAO also increased adipocyte area and decreased the CPT-1B expression in subcutaneous fat (P < 0.05). Taken together, TMAO supplementation regulated ileal microbial composition and acetate production, and regulated fat distribution and fatty acid composition in growing-finishing pigs. These results provide new insights for understanding the role of TMAO in humans and animals.
Collapse
Affiliation(s)
- Andong Zha
- Laboratory of Animal Nutritional Physiology and Metabolic Process, 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 410125, China
- University of Chinese Academy of Sciences, Beijing 100008, China
| | - Wanquan Li
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Jing Wang
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Ping Bai
- Yunnan Southwest Agriculture and Animal Husbandry Group Co., Ltd, Kunming 650224, China
| | - Ming Qi
- Laboratory of Animal Nutritional Physiology and Metabolic Process, 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 410125, China
- University of Chinese Academy of Sciences, Beijing 100008, China
| | - Peng Liao
- Laboratory of Animal Nutritional Physiology and Metabolic Process, 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 410125, China
| | - Bie Tan
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Yulong Yin
- Laboratory of Animal Nutritional Physiology and Metabolic Process, 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 410125, China
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
- University of Chinese Academy of Sciences, Beijing 100008, China
- Yunnan Southwest Agriculture and Animal Husbandry Group Co., Ltd, Kunming 650224, China
| |
Collapse
|
7
|
Wu J, Liu F, Sun J, Wei Q, Kang W, Wang F, Zhang C, Zhao M, Xu S, Han B. Toxic effects of acaricide fenazaquin on development, hemolymph metabolome, and gut microbiome of honeybee (Apis mellifera) larvae. CHEMOSPHERE 2024; 358:142207. [PMID: 38697560 DOI: 10.1016/j.chemosphere.2024.142207] [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: 02/18/2024] [Revised: 04/06/2024] [Accepted: 04/29/2024] [Indexed: 05/05/2024]
Abstract
Fenazaquin, a potent insecticide widely used to control phytophagous mites, has recently emerged as a potential solution for managing Varroa destructor mites in honeybees. However, the comprehensive impact of fenazaquin on honeybee health remains insufficiently understood. Our current study investigated the acute and chronic toxicity of fenazaquin to honeybee larvae, along with its influence on larval hemolymph metabolism and gut microbiota. Results showed that the acute median lethal dose (LD50) of fenazaquin for honeybee larvae was 1.786 μg/larva, and the chronic LD50 was 1.213 μg/larva. Although chronic exposure to low doses of fenazaquin exhibited no significant effect on larval development, increasing doses of fenazaquin resulted in significant increases in larval mortality, developmental time, and deformity rates. At the metabolic level, high doses of fenazaquin inhibited nucleotide, purine, and lipid metabolism pathways in the larval hemolymph, leading to energy metabolism disorders and physiological dysfunction. Furthermore, high doses of fenazaquin reduced gut microbial diversity and abundance, characterized by decreased relative abundance of functional gut bacterium Lactobacillus kunkeei and increased pathogenic bacterium Melissococcus plutonius. The disrupted gut microbiota, combined with the observed gut tissue damage, could potentially impair food digestion and nutrient absorption in the larvae. Our results provide valuable insights into the complex and diverse effects of fenazaquin on honeybee larvae, establishing an important theoretical basis for applying fenazaquin in beekeeping.
Collapse
Affiliation(s)
- Jiangli Wu
- State Key Laboratory of Resource Insects, Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Fengying Liu
- State Key Laboratory of Resource Insects, Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Jiajing Sun
- State Key Laboratory of Resource Insects, Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Qiaohong Wei
- State Key Laboratory of Resource Insects, Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Weipeng Kang
- State Key Laboratory of Resource Insects, Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Feng Wang
- Institute of Horticultural Research, Shanxi Academy of Agricultural Sciences, Shanxi Agricultural University, Taiyuan, 030031, China
| | - Chenhuan Zhang
- Zhejiang Provincial Key Laboratory of Biometrology and Inspection & Quarantine, College of Life Sciences, China Jiliang University, Hangzhou, 310018, China
| | - Meijiao Zhao
- State Key Laboratory of Resource Insects, Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Shufa Xu
- State Key Laboratory of Resource Insects, Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing, 100193, China.
| | - Bin Han
- State Key Laboratory of Resource Insects, Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing, 100193, China.
| |
Collapse
|
8
|
Yan Y, Li Q, Yang F, Shen L, Guo K, Zhou X. Chlorogenic acid ameliorates intestinal inflammation via miRNA-microbe axis in db/db mice. FASEB J 2024; 38:e23665. [PMID: 38780085 DOI: 10.1096/fj.202400382r] [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: 02/18/2024] [Revised: 04/18/2024] [Accepted: 04/29/2024] [Indexed: 05/25/2024]
Abstract
Chlorogenic acid improves diabetic symptoms, including inflammation, via the modulation of the gut microbiota. However, the mechanism by which the microbiota is regulated by chlorogenic acid remains unknown. In this study, we firstly explored the effects of chlorogenic acid on diabetic symptoms, colonic inflammation, microbiota composition, and microRNA (miRNA) expression in db/db mice. The results showed that chlorogenic acid decreased body weight, improved glucose tolerance and intestinal inflammation, altered gut microbiota composition, and upregulated the expression level of five miRNAs, including miRNA-668-3p, miRNA-467d-5p, miRNA-129-1-3p, miRNA-770-3p, and miRNA-666-5p in the colonic content. Interestingly, the levels of these five miRNAs were positively correlated with the abundance of Lactobacillus johnsonii. We then found that miRNA-129-1-3p and miRNA-666-5p promoted the growth of L. johnsonii. Importantly, miRNA-129-1-3p mimicked the effects of chlorogenic acid on diabetic symptoms and colonic inflammation in db/db mice. Furthermore, L. johnsonii exerted beneficial effects on db/db mice similar to those of chlorogenic acid. In conclusion, chlorogenic acid regulated the gut microbiota composition via affecting miRNA expression and ameliorated intestinal inflammation via the miRNA-microbe axis in db/db mice.
Collapse
Affiliation(s)
- Yongwang Yan
- Pharmaceutical College, Changsha Health Vocational College, Changsha, China
| | - Qing Li
- Pharmaceutical College, Changsha Health Vocational College, Changsha, China
- Department of Pathology, Changsha Health Vocational College, Changsha, China
| | - Fengluan Yang
- Obstetrics and Gynecology, 921 Hospital of the Chinese People's Liberation Army, Changsha, China
| | - Ling Shen
- Pharmaceutical College, Changsha Health Vocational College, Changsha, China
| | - Kangxiao Guo
- Pharmaceutical College, Changsha Health Vocational College, Changsha, China
- National Engineering Laboratory for Rice and By-Product Deep Processing, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha, China
| | - Xu Zhou
- Department of Spleen, Stomach and Liver Diseases, Affiliated Hospital of Hunan Academy of Traditional Chinese Medicine, Changsha, China
| |
Collapse
|
9
|
Zhan F, Song W, Fan Y, Wang F, Wang Q. Cucurbitacin E Alleviates Colonic Barrier Function Impairment and Inflammation Response and Improves Microbial Composition on Experimental Colitis Models. J Inflamm Res 2024; 17:2745-2756. [PMID: 38737108 PMCID: PMC11086439 DOI: 10.2147/jir.s456353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Accepted: 04/16/2024] [Indexed: 05/14/2024] Open
Abstract
Purpose Cucurbitacins, which are found in a variety of medicinal plants, vegetables and fruits, were known for their diverse pharmacological and biological activities, including anticancer, anti-oxidative and anti-inflammatory effects. Cucurbitacin E, one of the major cucurbitacins, was recently proved to inhibit inflammatory response. Methods To explore the therapeutic effects of cucurbitacin E on colitis and the underlying mechanisms, male mice drunk water containing 2.5% dextran sulfate sodium (DSS) to establish colitis model and administrated with cucurbitacin E during and after DSS treatment. The disease activity index was scored and colonic histological damage was observed. Intestinal tight junction and inflammatory response were determined. 16S rRNA and transcriptome sequencing were performed to analyze gut microbiota composition and gene expression, respectively. Results We found that cucurbitacin E alleviated DSS-induced body weight loss and impaired colonic morphology. Cucurbitacin E decreased the expression of inflammatory cytokines and cell apoptosis, and maintained barrier function. Additionally, cucurbitacin E retrieved DSS-induced alterations in the bacterial community composition. Furthermore, a variety of differentially expressed genes (DEGs) caused by cucurbitacin E were enriched in several pathways including the NFκB and TNF signaling pathways as well as in Th17 cell differentiation. There was a close relationship between DEGs and bacteria such as Escherichia-Shigella and Muribaculaceae. Conclusion Our results revealed that cucurbitacin E may exert protective effects on colitis via modulating inflammatory response, microbiota composition and host gene expression. Our study supports the therapeutic potential of cucurbitacin E in colitis and indicates that gut microbes are potentially therapeutic targets.
Collapse
Affiliation(s)
- Fengxia Zhan
- Department of Clinical Laboratory, Hospital of Shandong University, Jinan, 250100, People’s Republic of China
| | - Wei Song
- Key Laboratory of Marine Eco-Environmental Science and Technology, First Institute of Oceanography, Ministry of Natural Resources, Qingdao, 266061, People’s Republic of China
| | - Yong Fan
- Qingdao Mental Health Center, Qingdao University, Qingdao, People’s Republic of China
| | - Fangjian Wang
- Department of Clinical Laboratory, Hospital of Shandong University, Jinan, 250100, People’s Republic of China
| | - Qian Wang
- Department of Clinical Laboratory, Qilu Hospital, Shandong University, Jinan, 250012, People’s Republic of China
- Department of Clinical Laboratory, Qilu Hospital (Qingdao), Shandong University, Qingdao, 266035, People’s Republic of China
| |
Collapse
|
10
|
Zhu J, Fan Y, Yang S, Qin M, Chen X, Luo J, Chen T, Sun J, Zhang Y, Xi Q. Oral delivery of miR-146a-5p overexpression plasmid-loaded Pickering double emulsion modulates intestinal inflammation and the gut microbe. Int J Biol Macromol 2024; 261:129733. [PMID: 38307433 DOI: 10.1016/j.ijbiomac.2024.129733] [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: 06/05/2023] [Revised: 01/22/2024] [Accepted: 01/22/2024] [Indexed: 02/04/2024]
Abstract
The function of miRNAs in intestinal inflammatory injury regulation has been studied extensively. However, the targeted delivery of these functional nucleic acid molecules to specific organs through encapsulation carriers and exerting their functional effects remain critical challenges for further research. Here, we constructed miR-146a-5p overexpression plasmid and validated the anti-inflammatory properties in the cell model. Then, the plasmid was encapsulated by the Pickering double emulsion system to investigate the role of Pickering double emulsion system in LPS-induced acute intestinal inflammatory injury. The results showed that the Pickering double emulsion system could effectively protect the integrity of plasmids in the intestinal tract, alleviate intestinal inflammatory injury, and upregulate the relative abundance of Lactobacillus reuteri. Mechanically, in vivo and in vitro experiments have shown that miR-146a-5p inhibits TLR4/NF-κB pathway to alleviate intestinal inflammation. In addition, miR-146a-5p can also regulate intestinal homeostasis by targeting the RNA polymerase sigma factor RpoD and α-galactosidase A, thereby affecting the growth of Lactobacillus reuteri. Above all, this study reveals a potential mechanism for miR-146a-5p to treat intestinal inflammation and provides a new delivery strategy for miRNAs to regulate intestinal homeostasis.
Collapse
Affiliation(s)
- Jiahao Zhu
- State Key Laboratory of Livestock and Poultry Breeding, Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, No. 483 Wushan Road, Guangzhou 510642, China
| | - Yaotian Fan
- State Key Laboratory of Livestock and Poultry Breeding, Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, No. 483 Wushan Road, Guangzhou 510642, China
| | - Songfeng Yang
- State Key Laboratory of Livestock and Poultry Breeding, Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, No. 483 Wushan Road, Guangzhou 510642, China
| | - Mengran Qin
- State Key Laboratory of Livestock and Poultry Breeding, Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, No. 483 Wushan Road, Guangzhou 510642, China
| | - Xingping Chen
- Key Laboratory of Animal Nutrition in Jiangxi Province, Jiangxi Agricultural University, Nanchang, China
| | - Junyi Luo
- State Key Laboratory of Livestock and Poultry Breeding, Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, No. 483 Wushan Road, Guangzhou 510642, China
| | - Ting Chen
- State Key Laboratory of Livestock and Poultry Breeding, Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, No. 483 Wushan Road, Guangzhou 510642, China
| | - Jiajie Sun
- State Key Laboratory of Livestock and Poultry Breeding, Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, No. 483 Wushan Road, Guangzhou 510642, China
| | - Yongliang Zhang
- State Key Laboratory of Livestock and Poultry Breeding, Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, No. 483 Wushan Road, Guangzhou 510642, China
| | - Qianyun Xi
- State Key Laboratory of Livestock and Poultry Breeding, Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, No. 483 Wushan Road, Guangzhou 510642, China.
| |
Collapse
|
11
|
Tang W, Xiang X, Wang H, Zhou W, He L, Yin Y, Li T. Zinc lactate alleviates oxidative stress by modulating crosstalk between constitutive androstane receptor signaling pathway and gut microbiota profile in weaned piglets. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2024; 16:23-33. [PMID: 38131030 PMCID: PMC10730354 DOI: 10.1016/j.aninu.2023.10.001] [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: 04/26/2023] [Revised: 10/05/2023] [Accepted: 10/10/2023] [Indexed: 12/23/2023]
Abstract
This study aimed to determine the regulatory mechanism of dietary zinc lactate (ZL) supplementation on intestinal oxidative stress damage in a paraquat (PQ)-induced piglet model. Twenty-eight piglets (mean body weight 9.51 ± 0.23 kg) weaned at 28 d of age were randomly divided into control, ZL, PQ, and ZL + PQ groups (n = 7 in each group). The ZL-supplemented diet had little effect on growth performance under normal physiological conditions. However, under PQ challenge, ZL supplementation significantly improved average daily gain (P < 0.05) and reduced the frequency of diarrhea. ZL improved intestinal morphology and ultrastructure by significantly increasing the expression level of the jejunal tight junction protein, zonula occludens-1 (ZO-1) (P < 0.05), and intestinal zinc transport and absorption in PQ-induced piglets, which reduced intestinal permeability. ZL supplementation also enhanced the expression of antioxidant and anti-inflammatory factor-related genes and decreased inflammatory cytokine expression and secretion in PQ-induced piglets. Furthermore, ZL treatment significantly inhibited the activation of constitutive androstane receptor (CAR) signaling (P < 0.01) in PQ-induced piglets and altered the structure of the gut microbiota, especially by significantly increasing the abundance of beneficial gut microbes, including UCG_002, Ruminococcus, Rikenellaceae_RC9_gut_group, Christensenellaceae_R_7_group, Treponema, unclassified_Christensenellaceae, and unclassified_Erysipelotrichaceae (P < 0.05). These data reveal that pre-administration of ZL to piglets can suppress intestinal oxidative stress by improving antioxidant and anti-inflammatory capacity and regulating the crosstalk between CAR signaling and gut microbiota.
Collapse
Affiliation(s)
- Wenjie Tang
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
- Sichuan Academy of Animal Sciences, Animal Breeding and Genetics Key Laboratory of Sichuan Province, Chengdu 610066, China
- Livestock and Poultry Biological Products Key Laboratory of Sichuan Province, Sichuan Animtech Feed Co., Ltd, Chengdu 610000, China
| | - Xuan Xiang
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, CAS 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 410125, China
- College of Advanced Agricultural Science, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Houfu Wang
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, CAS 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 410125, China
| | - Wentao Zhou
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, CAS 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 410125, China
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, Changsha 410081, China
| | - Liuqin He
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, CAS 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 410125, China
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, Changsha 410081, China
| | - Yulong Yin
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, CAS 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 410125, China
- College of Advanced Agricultural Science, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Tiejun Li
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, CAS 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 410125, China
- College of Advanced Agricultural Science, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
12
|
Wang Y, Li Y, Lv L, Zhu L, Hong L, Wang X, Zhang Y, Wang X, Diao H. Faecal hsa-miR-7704 inhibits the growth and adhesion of Bifidobacterium longum by suppressing ProB and aggravates hepatic encephalopathy. NPJ Biofilms Microbiomes 2024; 10:13. [PMID: 38396001 PMCID: PMC10891095 DOI: 10.1038/s41522-024-00487-8] [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: 07/29/2023] [Accepted: 02/12/2024] [Indexed: 02/25/2024] Open
Abstract
Both gut microbiome and microRNAs (miRNAs) play a role in the development of hepatic encephalopathy (HE). However, the functional link between the microbiome and host-derived miRNAs in faeces remains poorly understood. In the present study, patients with HE had an altered gut microbiome and faecal miRNAs compared with patients with chronic hepatitis B. Transferring faeces and faecal miRNAs from patients with HE to the recipient mice aggravated thioacetamide-induced HE. Oral gavage of hsa-miR-7704, a host-derived miRNA highly enriched in faeces from patients with HE, aggravated HE in mice in a microbiome-dependent manner. Mechanistically, hsa-miR-7704 inhibited the growth and adhesion of Bifidobacterium longum by suppressing proB. B. longum and its metabolite acetate alleviated HE by inhibiting microglial activation and ammonia production. Our findings reveal the role of miRNA-microbiome axis in HE and suggest that faecal hsa-miR-7704 are potential regulators of HE progression.
Collapse
Affiliation(s)
- Yuchong Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Yuyu Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Longxian Lv
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Liying Zhu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Institute of Food Science, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310021, China
| | - Liang Hong
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Xueyao Wang
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, Shandong Province, China
| | - Yu Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Xin Wang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Institute of Food Science, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310021, China
| | - Hongyan Diao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.
| |
Collapse
|
13
|
Liu Y, Chen J, Wang S, Zhou X. Effects of two concentrations of dietary tribasic zinc sulfate on growth performance, gut morphology, and zinc transporter expression levels in pigs. Anim Biotechnol 2023; 34:2910-2916. [PMID: 36137171 DOI: 10.1080/10495398.2022.2125402] [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: 11/01/2022]
Abstract
Tribasic zinc sulfate (TBZ) is insoluble in water and chemically less active than zinc sulfate, making it more suitable to be used in pig diet. To investigate the effects of TBZ on the growth performance, gut morphology, and zinc transporter expression levels, we performed a single-factor experiment and 168 pigs were allocated to three groups with seven pens per treatment. Pigs were either fed a basal diet without zinc supplementation (control group), or a basal diet supplemented with TBZ at 100 mg/kg diet (LTBZ group) or 1000 mg/kg diet (HTBZ group). We found that daily weight gain and feed intake were higher in the LTBZ group than in the HTBZ and control groups. The pigs in the LTBZ group had a higher villus height and villus height/crypt depth ratio when compared with other pigs. Moreover, the pigs in the LTBZ group exhibited higher mRNA expression levels of solute carrier family 39 and lower expression levels of solute carrier family 30 than those fed the HTBZ-supplemented diet. Together, these results indicate that TBZ may potentially be used as a dietary zinc source for young growing pigs and that dietary supplementation with LTBZ benefits growth performance and gut morphology.
Collapse
Affiliation(s)
- Yonghui Liu
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, College of Life Sciences, Hunan Normal University, Changsha, China
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
| | - Juan Chen
- Changsha Xingjia Biotech Co., Ltd, Changsha, China
| | | | - Xihong Zhou
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
| |
Collapse
|
14
|
Zhou X, He Y, Chen J, Xiong X, Yin J, Liang J, Peng C, Huang C, Guan G, Yin Y. Colonic phosphocholine is correlated with Candida tropicalis and promotes diarrhea and pathogen clearance. NPJ Biofilms Microbiomes 2023; 9:62. [PMID: 37666845 PMCID: PMC10477305 DOI: 10.1038/s41522-023-00433-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 08/24/2023] [Indexed: 09/06/2023] Open
Abstract
Diarrhea is characterized by alterations in the gut microbiota, metabolites, and host response to these changes. Studies have focused on the role of commensal bacteria in diarrhea; however, the effect of fungi on its pathogenesis remains unexplored. Here, using post-weaned piglets with or without diarrhea, we found an unexpected decrease in the abundance of Candida tropicalis in diarrheal piglets. We also observed increased accumulation of reactive oxygen species (ROS) and the formation of neutrophil extracellular traps (NETs) in the colonic tissues of diarrheal piglets. Using dectin-1-knockout mice, we found that the over-accumulation of ROS killed C. tropicalis by promoting NET formation, which was dependent on dectin-1. The decreased abundance of C. tropicalis resulted in reduced phosphocholine consumption. Then, colonic phosphocholine accumulation drives water efflux by increasing cAMP levels by activating adenylyl cyclase, which promotes the clearance of pathogenic bacteria. Collectively, we demonstrated that phosphocholine is correlated with colonic C. tropicalis and promotes diarrhea and pathogen clearance. Our results suggest that mycobiota colonizing the colon might be involved in maintaining intestinal metabolic homeostasis through the consumption of certain metabolites.
Collapse
Affiliation(s)
- Xihong Zhou
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, the Chinese Academy of Sciences, Changsha, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
- School of Stomatology, Changsha Medical University, Changsha, China
| | - Yiwen He
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, the Chinese Academy of Sciences, Changsha, China
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Jingqing Chen
- Laboratory Animal Center of the Academy of Military Medical Sciences, Beijing, China
| | - Xia Xiong
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, the Chinese Academy of Sciences, Changsha, China.
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China.
- School of Stomatology, Changsha Medical University, Changsha, China.
| | - Jie Yin
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China.
| | - Jing Liang
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, the Chinese Academy of Sciences, Changsha, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Can Peng
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, the Chinese Academy of Sciences, Changsha, China
| | - Chunxia Huang
- School of Stomatology, Changsha Medical University, Changsha, China
| | - Guiping Guan
- College of Bioscience & Biotechnology, Hunan Agricultural University, Changsha, China
| | - Yulong Yin
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, the Chinese Academy of Sciences, Changsha, China.
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China.
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China.
| |
Collapse
|
15
|
Wang Z, Zeng B, Xue H, Liu C, Song W. Blidingia sp. extracts improve intestinal health and reduce diarrhoea in weanling piglets. J Anim Physiol Anim Nutr (Berl) 2023; 107:1198-1205. [PMID: 37203256 DOI: 10.1111/jpn.13832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 03/12/2023] [Accepted: 04/13/2023] [Indexed: 05/20/2023]
Abstract
Blidingia sp. is a prominent fouling green macroalga and we previously found that extracts from Blidingia sp. alleviated intestinal inflammation in mice challenged with lipopolysaccharides. However, whether these extracts are effective in weanling piglets remains unknown. In the present study, Blidingia sp. extracts were supplemented in the diet and their effects on growth performance, incidence of diarrhoea and intestinal function in weanling piglets were explored. The results showed that diets supplemented with 0.1% or 0.5% Blidingia sp. extract significantly increased average daily body weight gain and feed intake in weanling piglets. Meanwhile, piglets supplemented with 0.5% Blidingia sp. extract showed decreased incidence of diarrhoea as well as reduced fecal water and Na+ content. Furthermore, the diet supplemented with 0.5% Blidingia sp. extracts improved intestinal morphology, as indicated by the results of hematoxylin and eosin staining. Diet supplemented with 0.5% Blidingia sp. extracts also improved tight junction function, as indicated by increased expression of Occludin, Claudin-1 and Zonula occludens-1, and alleviated the inflammatory response, as indicated by decreased tumor necrosis factor-α and interleukin-6 (IL6) contents and increased IL10 levels. Taken together, our results showed that Blidingia sp. extracts had beneficial effects in weanling piglets and we suggest that Blidingia sp. extracts could be potentially used as an additive for piglets.
Collapse
Affiliation(s)
- Zongling Wang
- Key Laboratory of Marine Eco-Environmental Science and Technology, First Institute of Oceanography, Ministry of Natural Resources, Qingdao, China
- Laboratory of Marine Ecology and Environmental Science, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, China
| | - Boxin Zeng
- Key Laboratory of Marine Eco-Environmental Science and Technology, First Institute of Oceanography, Ministry of Natural Resources, Qingdao, China
- Laboratory of Marine Ecology and Environmental Science, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, China
| | - Haoran Xue
- Department of Clinical Laboratory, Qilu Hospital (Qingdao) of Shandong University, Qingdao, China
| | - Chunyan Liu
- Department of Integrated Traditional Chinese and Western Medicine, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Wei Song
- Key Laboratory of Marine Eco-Environmental Science and Technology, First Institute of Oceanography, Ministry of Natural Resources, Qingdao, China
- Laboratory of Marine Ecology and Environmental Science, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, China
| |
Collapse
|
16
|
Datta N, Johnson C, Kao D, Gurnani P, Alexander C, Polytarchou C, Monaghan TM. MicroRNA-based therapeutics for inflammatory disorders of the microbiota-gut-brain axis. Pharmacol Res 2023; 194:106870. [PMID: 37499702 DOI: 10.1016/j.phrs.2023.106870] [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: 03/11/2023] [Revised: 07/22/2023] [Accepted: 07/24/2023] [Indexed: 07/29/2023]
Abstract
An emerging but less explored shared pathophysiology across microbiota-gut-brain axis disorders is aberrant miRNA expression, which may represent novel therapeutic targets. miRNAs are small, endogenous non-coding RNAs that are important transcriptional repressors of gene expression. Most importantly, they regulate the integrity of the intestinal epithelial and blood-brain barriers and serve as an important communication channel between the gut microbiome and the host. A well-defined understanding of the mode of action, therapeutic strategies and delivery mechanisms of miRNAs is pivotal in translating the clinical applications of miRNA-based therapeutics. Accumulating evidence links disorders of the microbiota-gut-brain axis with a compromised gut-blood-brain-barrier, causing gut contents such as immune cells and microbiota to enter the bloodstream leading to low-grade systemic inflammation. This has the potential to affect all organs, including the brain, causing central inflammation and the development of neurodegenerative and neuropsychiatric diseases. In this review, we have examined in detail miRNA biogenesis, strategies for therapeutic application, delivery mechanisms, as well as their pathophysiology and clinical applications in inflammatory gut-brain disorders. The research data in this review was drawn from the following databases: PubMed, Google Scholar, and Clinicaltrials.gov. With increasing evidence of the pathophysiological importance for miRNAs in microbiota-gut-brain axis disorders, therapeutic targeting of cross-regulated miRNAs in these disorders displays potentially transformative and translational potential. Further preclinical research and human clinical trials are required to further advance this area of research.
Collapse
Affiliation(s)
- Neha Datta
- School of Medicine, University of Nottingham, Nottingham, UK
| | - Charlotte Johnson
- NIHR Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, UK; Nottingham Digestive Diseases Centre, School of Medicine, University of Nottingham, Nottingham, UK
| | - Dina Kao
- Division of Gastroenterology, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Pratik Gurnani
- Division of Molecular Therapeutics & Formulation, School of Pharmacy, University of Nottingham, Nottingham, UK
| | - Cameron Alexander
- Division of Molecular Therapeutics & Formulation, School of Pharmacy, University of Nottingham, Nottingham, UK
| | - Christos Polytarchou
- Department of Biosciences, John van Geest Cancer Research Centre, School of Science & Technology, Nottingham Trent University, Nottingham, UK.
| | - Tanya M Monaghan
- NIHR Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, UK; Nottingham Digestive Diseases Centre, School of Medicine, University of Nottingham, Nottingham, UK.
| |
Collapse
|
17
|
Zhang C, Liu H, Sun L, Wang Y, Chen X, Du J, Sjöling Å, Yao J, Wu S. An overview of host-derived molecules that interact with gut microbiota. IMETA 2023; 2:e88. [PMID: 38868433 PMCID: PMC10989792 DOI: 10.1002/imt2.88] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 01/17/2023] [Accepted: 01/21/2023] [Indexed: 06/14/2024]
Abstract
The gut microbiota comprises bacteria, archaea, fungi, protists, and viruses that live together and interact with each other and with host cells. A stable gut microbiota is vital for regulating host metabolism and maintaining body health, while a disturbed microbiota may induce different kinds of disease. In addition, diet is also considered to be the main factor that influences the gut microbiota. The host could shape the gut microbiota through other factors. Here, we reviewed the mechanisms that mediate host regulation on gut microbiota, involved in gut-derived molecules, including gut-derived immune system molecules (secretory immunoglobulin A, antimicrobial peptides, cytokines, cluster of differentiation 4+ effector T cell, and innate lymphoid cells), sources related to gut-derived mucosal molecules (carbon sources, nitrogen sources, oxygen sources, and electron respiratory acceptors), gut-derived exosomal noncoding RNA (ncRNAs) (microRNAs, circular RNA, and long ncRNA), and molecules derived from organs other than the gut (estrogen, androgen, neurohormones, bile acid, and lactic acid). This study provides a systemic overview for understanding the interplay between gut microbiota and host, a comprehensive source for potential ways to manipulate gut microbiota, and a solid foundation for future personalized treatment that utilizes gut microbiota.
Collapse
Affiliation(s)
- Chenguang Zhang
- College of Animal Science and TechnologyNorthwest A&F UniversityYanglingChina
| | - Huifeng Liu
- College of Animal Science and TechnologyNorthwest A&F UniversityYanglingChina
| | - Lei Sun
- Centre for Translational Microbiome Research, Department of Microbiology, Tumor and Cell BiologyKarolinska InstitutetStockholmSweden
| | - Yue Wang
- College of Animal Science and TechnologyNorthwest A&F UniversityYanglingChina
| | - Xiaodong Chen
- College of Animal Science and TechnologyNorthwest A&F UniversityYanglingChina
| | - Juan Du
- Centre for Translational Microbiome Research, Department of Microbiology, Tumor and Cell BiologyKarolinska InstitutetStockholmSweden
| | - Åsa Sjöling
- Centre for Translational Microbiome Research, Department of Microbiology, Tumor and Cell BiologyKarolinska InstitutetStockholmSweden
| | - Junhu Yao
- College of Animal Science and TechnologyNorthwest A&F UniversityYanglingChina
| | - Shengru Wu
- College of Animal Science and TechnologyNorthwest A&F UniversityYanglingChina
| |
Collapse
|
18
|
Liu S, Song Q, Zhang C, Li M, Li Z, Liu Y, Xu L, Xie X, Zhao L, Zhang R, Wang Q, Zeng G, Zhang Y, Zhang K. Saliva microbiome alterations in dental fluorosis population. J Oral Microbiol 2023; 15:2180927. [PMID: 36844898 PMCID: PMC9946311 DOI: 10.1080/20002297.2023.2180927] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2023] Open
Abstract
Background We aimed to explore saliva microbiome alterations in dental fluorosis population. Methods The prevalence of dental fluorosis was examined in 957 college students. Dean's fluorosis index was used to evaluate the dental fluorosis status. Changes in the composition of the salivary microbiome were assessed in a subset of these patients (100 healthy controls, 100 dental fluorosis patients). Results Dental fluorosis affected 47% of the student sample, and incidence was unrelated to gender. Compared with healthy controls, the microbiota of patients with dental fluorosis exhibited increased diversity, with increased abundance of Treponema lecithinolyticum, Vibrio metschnikovii, Cupriavidus pauculus, Pseudomonas, Pseudomonadaceae, Pseudomonadales, and decreased abundance of Streptococcus mutans, Streptococcus sanguinis, Gemella, and Staphylococcales. Function analyses showed increases in arginine biosynthesis in patients affected by dental fluorosis, together with reductions in amino sugar and nucleotide sugar metabolism, fructose and mannose metabolism, and starch and sucrose metabolism. Conclusions These results suggest that there are striking differences in salivary microbiome between healthy controls and dental fluorosis patients. Dental fluorosis may contribute to periodontitis and systemic lung diseases. There is a need for cohort studies to determine whether altering the salivary microbiota in dental fluorosis patients can alter the development of oral or systemic diseases.
Collapse
Affiliation(s)
- Shanshan Liu
- Department of Stomatology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China,Anhui Key Laboratory of Infection and Immunity, Bengbu Medical College, Bengbu, China,Department of Stomatology, Bengbu Medical College, Bengbu, China
| | - Qiangsheng Song
- Department of Stomatology, Huaiyuan county people’s Hospital, Bengbu, China
| | - Chenchen Zhang
- Department of Stomatology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China,Department of Stomatology, Bengbu Medical College, Bengbu, China
| | - Mengwan Li
- Department of Stomatology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Zhenzhen Li
- Department of Stomatology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Yudong Liu
- Department of Histology and Embryology, Bengbu Medical College, Bengbu, China
| | - Li Xu
- Department of Stomatology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Xiaofei Xie
- Department of Stomatology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Lili Zhao
- Department of Stomatology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Rongxiu Zhang
- Department of Stomatology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Qinglong Wang
- Department of Stomatology, Bengbu Medical College, Bengbu, China
| | - Guojin Zeng
- Department of Stomatology, Bengbu Medical College, Bengbu, China
| | - Yifan Zhang
- Department of Stomatology, Bengbu Medical College, Bengbu, China
| | - Kai Zhang
- Department of Stomatology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China,CONTACT Kai Zhang Department of Stomatology, The First Affiliated Hospital of Bengbu Medical College, 287 Chuang Huai Road, Bengbu233004, China
| |
Collapse
|
19
|
Yu Z, Chen J, Liu Y, Meng Q, Liu H, Yao Q, Song W, Ren X, Chen X. The role of potential probiotic strains Lactobacillus reuteri in various intestinal diseases: New roles for an old player. Front Microbiol 2023; 14:1095555. [PMID: 36819028 PMCID: PMC9932687 DOI: 10.3389/fmicb.2023.1095555] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 01/03/2023] [Indexed: 02/05/2023] Open
Abstract
Lactobacillus reuteri (L. reuteri), a type of Lactobacillus spp., is a gut symbiont that can colonize many mammals. Since it was first isolated in 1962, a multitude of research has been conducted to investigate its function and unique role in different diseases as an essential probiotic. Among these, the basic functions, beneficial effects, and underlying mechanisms of L. reuteri have been noticed and understood profoundly in intestinal diseases. The origins of L. reuteri strains are diverse, with humans, rats, and piglets being the most common. With numerous L. reuteri strains playing significant roles in different intestinal diseases, DSM 17938 is the most widely used in humans, especially in children. The mechanisms by which L. reuteri improves intestinal disorders include protecting the gut barrier, suppressing inflammation and the immune response, regulating the gut microbiota and its metabolism, and inhibiting oxidative stress. While a growing body of studies focused on L. reuteri, there are still many unknowns concerning its curative effects, clinical safety, and precise mechanisms. In this review, we initially interpreted the basic functions of L. reuteri and its related metabolites. Then, we comprehensively summarized its functions in different intestinal diseases, including inflammatory bowel disease, colorectal cancer, infection-associated bowel diseases, and pediatric intestinal disorders. We also highlighted some important molecules in relation to the underlying mechanisms. In conclusion, L. reuteri has the potential to exert a beneficial impact on intestinal diseases, which should be further explored to obtain better clinical application and therapeutic effects.
Collapse
Affiliation(s)
- Zihan Yu
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China,Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, China
| | - Jihua Chen
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China,Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, China
| | - Yaxin Liu
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China,Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, China
| | - Qingguo Meng
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China,Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, China
| | - Hang Liu
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China,Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, China
| | - Qinyan Yao
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China,Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, China
| | - Wenxuan Song
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China,Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiangfeng Ren
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China,Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, China
| | - Xin Chen
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China,Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, China,*Correspondence: Xin Chen ✉
| |
Collapse
|
20
|
Wang S, Wu S, Zhang Y, Chen J, Zhou X. Effects of Different Levels of Organic Trace Minerals on Oxidative Status and Intestinal Function in Weanling Piglets. Biol Trace Elem Res 2023; 201:720-727. [PMID: 35229256 DOI: 10.1007/s12011-022-03174-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 02/20/2022] [Indexed: 01/21/2023]
Abstract
An experiment was conducted to compare the effects of replacing inorganic trace minerals (ITM) with different levels of complex organic trace minerals (OTM) on the growth performance, oxidative status, and intestinal function of piglets. Weanling piglets were assigned to five groups: a control group fed a basal diet supplemented with inorganic trace minerals and the other four groups fed basal diets supplemented with different levels of OTMs. The results showed that diets supplemented with 50 ppm Fe, 30 ppm Zn, 15 ppm Mn, and 0.2 ppm Se from OTM (L-OTM), or with 75 ppm Fe, 45 ppm Zn, 22.5 ppm Mn, and 0.3 ppm Se from OTM (M-OTM) significantly decreased the diarrhea ratio in the piglets compared with those supplemented with 100 ppm Fe, 90 ppm Zn, 40 ppm Mn, and 0.4 ppm Se from ITM. Compared with those in the ITM group, the piglets in the M-OTM group had significantly higher serum CuZnSOD, MnSOD, and GSH-Px levels. Moreover, piglets in the L-OTM and M-OTM groups had higher Sod and Gpx gene expression than those in the ITM group. Additionally, piglets in the L-OTM and M-OTM groups had significantly higher villus height than those in the ITM group, and the M-OTM group piglets had lower serum diamine oxidase content and higher ileal ZO-1 and occludin protein expression levels than those in the ITM group. These results indicate that replacing dietary ITMs with OTMs could decrease diarrhea occurrence and improve the oxidative status and intestinal barrier function in weanling piglets.
Collapse
Affiliation(s)
- Shangchu Wang
- Changsha Xinjia Bio-Engineering Co., Ltd., Changsha, 410000, China
| | - Shujun Wu
- Changsha Xinjia Bio-Engineering Co., Ltd., Changsha, 410000, China
| | - Yawei Zhang
- Changsha Xinjia Bio-Engineering Co., Ltd., Changsha, 410000, China
| | - Juan Chen
- Changsha Xinjia Bio-Engineering Co., Ltd., Changsha, 410000, China
| | - Xihong Zhou
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China.
| |
Collapse
|
21
|
He Y, Liang J, Liu Y, Zhou X, Peng C, Long C, Huang P, Feng J, Zhang Z. Combined supplementation with Lactobacillus sp. and Bifidobacterium thermacidophilum isolated from Tibetan pigs improves growth performance, immunity, and microbiota composition in weaned piglets. J Anim Sci 2023; 101:skad220. [PMID: 37358243 PMCID: PMC10347973 DOI: 10.1093/jas/skad220] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 06/23/2023] [Indexed: 06/27/2023] Open
Abstract
Probiotics, such as Lactobacillus and Bifidobacterium, promote growth in piglets by modulating gut microbiota composition and improving the host immune system. A strain of Lactobacillus sp. and Bifidobacterium thermacidophilum were previously isolated from fresh feces of Tibetan pigs. The effects of these isolated strains on growth performance, intestinal morphology, immunity, microbiota composition, and their metabolites were evaluated in weaned piglets. Thirty crossbred piglets were selected and fed either a basal diet (CON), a basal diet supplemented with aureomycin (ANT), or a basal diet supplemented with Lactobacillus sp. and B. thermacidophilum (LB) for 28 d. The piglets in the ANT and LB groups had significantly higher body weight gain than those in the CON group (P < 0.05). Piglets in the ANT and LB groups had regularly arranged villi and microvilli in the small intestine. Furthermore, they had improved immune function, as indicated by decreased serum concentrations of inflammatory cytokines (P < 0.05), improved components of immune cells in the blood, mesenteric lymph nodes, and spleen. Additionally, metagenomic sequencing indicated a significant shift in cecal bacterial composition and alterations in microbiota functional profiles following Lactobacillus sp. and B. thermacidophilum supplementation. Metabolomic results revealed that the metabolites were also altered, and Kyoto Encyclopedia of Genes and Genomes analysis revealed that several significantly altered metabolites were enriched in glycerophospholipid and cholesterol metabolism (P < 0.05). Furthermore, correlation analysis showed that several bacterial members were closely related to the alterations in metabolites, including Bacteroides sp., which were negatively correlated with triglyceride (16:0/18:0/20:4[5Z,8Z,11Z,14Z]), the metabolite that owned the highest variable importance of projection scores. Collectively, our findings suggest that combined supplementation with Lactobacillus sp. and B. thermacidophilum significantly improved the growth performance, immunity, and microbiota composition in weaned piglets, making them prospective alternatives to antibiotics in swine production.
Collapse
Affiliation(s)
- Yiwen He
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, College of Life Sciences, Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Hunan Normal University, Changsha 410081, China
| | - Jing Liang
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
| | - Yonghui Liu
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, College of Life Sciences, Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Hunan Normal University, Changsha 410081, China
| | - Xihong Zhou
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
| | - Can Peng
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
| | - Ciming Long
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
| | - Pan Huang
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
| | - Jie Feng
- Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Zhigang Zhang
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Life Sciences, Yunnan University, Kunming 650091, China
| |
Collapse
|
22
|
Ma LT, Lian JX, Bai Y, Shang MJ, Zhang ZZ, Wu FF, Chen J, Meng XB, Zheng J, Li T, Li YQ, Wang JJ. Adeno-associated virus vector intraperitoneal injection induces colonic mucosa and submucosa transduction and alters the diversity and composition of the faecal microbiota in rats. Front Cell Infect Microbiol 2022; 12:1028380. [PMID: 36619753 PMCID: PMC9813966 DOI: 10.3389/fcimb.2022.1028380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 11/30/2022] [Indexed: 12/24/2022] Open
Abstract
Background Viral vector technology, especially recombinant adeno-associated virus vector (rAAV) technology, has shown great promise in preclinical research for clinical applications. Several studies have confirmed that rAAV can successfully transduce the enteric nervous system (ENS), and rAAV gene therapy has been approved by the Food and Drug Administration (FDA) for the treatment of the early childhood blindness disease Leber congenital amaurosis and spinal muscular atrophy (SMA). However, until now, it has not been possible to determine the effect of AAV9 on intestinal microbiota. Methods We examined the efficiency of AAV9-mediated ascending colon, transverse colon and descending colon transduction through intraperitoneal (IP) injection, performed 16S rRNA gene amplicon sequencing and analysed specific faecal microbial signatures following AAV9 IP injection via bioinformatics methods in Sprague-Dawley (SD) rats. Results Our results showed (1) efficient transduction of the mucosa and submucosa of the ascending, transverse, and descending colon following AAV9 IP injection; (2) a decreased alpha diversity and an altered overall microbial composition following AAV9 IP injection; (3) significant enrichments in a total of 5 phyla, 10 classes, 13 orders, 15 families, 29 genera, and 230 OTUs following AAV9 IP injection; and (4) AAV9 can significantly upregulate the relative abundance of anaerobic microbiota which is one of the seven high-level phenotypes that BugBase could predict. Conclusion In summary, these data show that IP injection of AAV9 can successfully induce the transduction of the colonic mucosa and submucosa and alter the diversity and composition of the faecal microbiota in rats.
Collapse
Affiliation(s)
- Li-Tian Ma
- Department of Gastroenterology, Tangdu Hospital, Air Force Medical University, Xi’an, China,Department of Traditional Chinese Medicine, Tangdu Hospital, Air Force Medical University, Xi’an, China
| | - Jing-Xuan Lian
- Department of Endocrinology, Xijing Hospital, Air Force Medical University, Xi’an, China
| | - Yang Bai
- Department of Neurosurgery, General Hospital of Northern Theater Command, Shenyang, China
| | - Meng-Juan Shang
- Department of Radiation Biology, Faculty of Preventive Medicine, Fourth Military Medical University, Xi'an, ShaanXi, China
| | - Zhe-Zhe Zhang
- Department of Traditional Chinese Medicine, Tangdu Hospital, Air Force Medical University, Xi’an, China
| | - Fei-Fei Wu
- National Demonstration Center for Experimental Preclinical Medicine Education, Air Force Medical University, Xi’an, China
| | - Jing Chen
- Department of Anatomy, Histology and Embryology & K. K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, China
| | - Xian-Bo Meng
- Department of Gastroenterology, Tangdu Hospital, Air Force Medical University, Xi’an, China
| | - Jin Zheng
- Department of Traditional Chinese Medicine, Tangdu Hospital, Air Force Medical University, Xi’an, China
| | - Tian Li
- School of Basic Medicine, Fourth Military Medical University, Xi'an, China,*Correspondence: Jing-Jie Wang, ; Yun-Qing Li, ; Tian Li,
| | - Yun-Qing Li
- Department of Anatomy, Histology and Embryology & K. K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, China,Key Laboratory of Brain Science Research and Transformation in Tropical Environment of Hainan Province, Haikou, China,Department of Anatomy, College of Basic Medicine, Dali University, Dali, China,*Correspondence: Jing-Jie Wang, ; Yun-Qing Li, ; Tian Li,
| | - Jing-Jie Wang
- Department of Gastroenterology, Tangdu Hospital, Air Force Medical University, Xi’an, China,*Correspondence: Jing-Jie Wang, ; Yun-Qing Li, ; Tian Li,
| |
Collapse
|
23
|
Dhuppar S, Murugaiyan G. miRNA effects on gut homeostasis: therapeutic implications for inflammatory bowel disease. Trends Immunol 2022; 43:917-931. [PMID: 36220689 PMCID: PMC9617792 DOI: 10.1016/j.it.2022.09.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 09/05/2022] [Accepted: 09/11/2022] [Indexed: 01/12/2023]
Abstract
Inflammatory bowel disease (IBD) spans a range of chronic conditions affecting the gastrointestinal (GI) tract, which are marked by intermittent flare-ups and remissions. IBD results from microbial dysbiosis or a defective mucosal barrier in the gut that triggers an inappropriate immune response in a genetically susceptible person, altering the immune-microbiome axis. In this review, we discuss the regulatory roles of miRNAs, small noncoding RNAs with gene regulatory functions, in the stability and maintenance of the gut immune-microbiome axis, and detail the challenges and recent advances in the use of miRNAs as putative therapeutic agents for treating IBD.
Collapse
Affiliation(s)
- Shivnarayan Dhuppar
- Division of Hematology, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Current address: Centre for Business Innovation, The Indian School of Business, Hyderabad 500111, India
| | - Gopal Murugaiyan
- Ann Romney Center for Neurological Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
24
|
Fan Y, Qin M, Zhu J, Chen X, Luo J, Chen T, Sun J, Zhang Y, Xi Q. MicroRNA sensing and regulating microbiota-host crosstalk via diet motivation. Crit Rev Food Sci Nutr 2022; 64:4116-4133. [PMID: 36287029 DOI: 10.1080/10408398.2022.2139220] [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: 11/03/2022]
Abstract
Accumulating evidence has demonstrated that diet-derived gut microbiota participates in the regulation of host metabolism and becomes the foundation for precision-based nutritional interventions and the biomarker for potential individual dietary recommendations. However, the specific mechanism of the gut microbiota-host crosstalk remains unclear. Recent studies have identified that noncoding RNAs, as important elements in the regulation of the initiation and termination of gene expression, mediate microbiota-host communication. Besides, the cross-kingdom regulation of non-host derived microRNAs also influence microbiota-host crosstalk via diet motivation. Hence, understanding the relationship between gut microbiota, miRNAs, and host metabolism is indispensable to revealing individual differences in dietary motivation and providing targeted recommendations and strategies. In this review, we first present an overview of the interaction between diet, host genetics, and gut microbiota and collected some latest research associated with microRNAs modulated gut microbiota and intestinal homeostasis. Then, specifically described the possible molecular mechanisms of microRNAs in sensing and regulating gut microbiota-host crosstalk. Lastly, summarized the prospect of microRNAs as biomarkers in disease diagnosis, and the disadvantages of microRNAs in regulating gut microbiota-host crosstalk. We speculated that microRNAs could become potential novel circulating biomarkers for personalized dietary strategies to achieve precise nutrition in future clinical research implications.
Collapse
Affiliation(s)
- Yaotian Fan
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Mengran Qin
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Jiahao Zhu
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Xingping Chen
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
- Key Laboratory of Animal Nutrition in Jiangxi Province, Jiangxi Agricultural University, Nanchang, China
| | - Junyi Luo
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Ting Chen
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Jiajie Sun
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Yongliang Zhang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Qianyun Xi
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| |
Collapse
|
25
|
Cui X, Chen J, Yang Y. Administration of selenomethionine in combination with serine benefits diabetes via gut microbiota. Front Microbiol 2022; 13:1007814. [PMID: 36312938 PMCID: PMC9597302 DOI: 10.3389/fmicb.2022.1007814] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 08/31/2022] [Indexed: 11/29/2022] Open
Abstract
Either selenium or serine could modulate glucose homeostasis, however, whether there are synergistic effects of selenium with serine on diabetes remains to be unknown. In the present study, eight male db/m mice were used as a control, and 24 male diabetic db/db mice were either orally gavaged with PBS, or with selenomethionine alone, or with both selenomethionine and serine, to investigate the effects of selenomethionine and serine on body weight and glucose level. Furthermore, intestinal microbiota composition was analyzed and fecal microbiota transplantation (FMT) was performed to explore whether microbes mediate the beneficial effects of selenomethionine and serine. The results showed that administration of selenomethionine decreased body weight, adipose tissue weight and serum glucose level in db/db diabetic mice. Importantly, administration of selenomethionine in combination with serine exerted better effects than selenomethionine alone did. Furthermore, a combined administration of selenomethionine and serine restored the microbial composition in diabetic mice. Corynebacterium glutamicum, Bifidobacterium pseudolongum, and Aerococcus urinaeequi were significantly decreased, whereas Lactobacillus murinus was increased in mice in the selenomethionine group and selenomethionine in combination with serine group, when compared with those in the db/db group. FMT decreased body weight and glucose level in db/db mice, further indicating that microbes play critical roles in the beneficial effects of selenomethionine and serine. Thus, we concluded that administration of selenomethionine in combination with serine benefits diabetes via gut microbes. Our results suggested that the synergic application of selenomethionine and serine could be potentially used for the treatment of diabetes.
Collapse
Affiliation(s)
- Xiaoyan Cui
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Jingqing Chen
- Laboratory Animal Center of the Academy of Military Medical Sciences, Beijing, China
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, China
| | - Yuexi Yang
- School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, China
- *Correspondence: Yuexi Yang,
| |
Collapse
|
26
|
Jellyfish Collagen Hydrolysate Alleviates Inflammation and Oxidative Stress and Improves Gut Microbe Composition in High-Fat Diet-Fed Mice. Mediators Inflamm 2022; 2022:5628702. [PMID: 35979013 PMCID: PMC9377926 DOI: 10.1155/2022/5628702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 07/17/2022] [Accepted: 07/22/2022] [Indexed: 11/17/2022] Open
Abstract
The collagen from jellyfish has many beneficial effects, including antioxidant, anti-inflammatory and immune-modulatory activities. However, whether jellyfish collagen hydrolysate (JCH) has any effects on high-fat diet-induced obesity remains unknown. Consequently, we in the present study orally administrated JCH in high-fat diet-fed mice to explore its effects on body weight gain, inflammatory and oxidative status, and cecum microbe composition. The results showed that oral administration of JCH prevented the body weight gain in high-fat diet-treated mice. Meanwhile, glucose, triglycerides, and total cholesterol level in serum were maintained by JCH administration. Furthermore, JCH administration alleviated oxidative stress by increasing the GSH content and decreasing the level reactive oxygen species in the liver and improved inflammatory response by decreasing the expression of TNF-α, IL-1β, and IL-8 gene in the liver and ileum. Importantly, JCH administration helps recover the alteration of microbiota composition induced by high-fat diet, and the genus Romboutsia may critically involve in the beneficial effects of JCH administration. In conclusion, our results indicated that JCH could be potentially used for the prevention and treatment of diet-induced obesity.
Collapse
|
27
|
Li Z, Ma S, Wang X, Wang Y, Yan R, Wang J, Xu Z, Wang S, Feng Y, Wang J, Mei Q, Yang P, Liu L. Pharmacokinetic and gut microbiota analyses revealed the effect of Lactobacillus acidophilus on the metabolism of Olsalazine in ulcerative colitis rats. Eur J Pharm Sci 2022; 175:106235. [PMID: 35697287 DOI: 10.1016/j.ejps.2022.106235] [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: 03/20/2022] [Revised: 05/21/2022] [Accepted: 06/09/2022] [Indexed: 11/03/2022]
Abstract
Olsalazine is a typical 5-aminosalicylic acid (5-ASA) drug that depends on gut microbiota to liberate its anti-inflammatory moiety 5-ASA in the treatment of ulcerative colitis (UC). In recent decades, 5-ASA drugs combined with probiotics have achieved a better effective treatment for UC. Mechanisms of combination therapy have been widely discussed from a pharmacodynamic perspective. However, it is still unclear whether the better therapeutic efficacy of combination therapy was made by changing the metabolism of 5-ASA drugs in the colon under the regulation of probiotics. In the present study, combined with pharmacokinetic and gut microbiota analyses, we systematically evaluated the potential effect of Lactobacillus acidophilus (L. acidophilus) on the metabolism of Olsalazine at three levels (pharmacokinetic characteristics, metabolic microbiota, and metabolic enzymes) to offer some insights into this issue. As pharmacokinetic results showed, L. acidophilus barely had an influence on the pharmacokinetic parameters of Olsalazine, 5-ASA, and N-Ac-5-ASA. Notably, the colonic exposure of 5-ASA was not affected by L. acidophilus. Gut microbiota results also illustrated that L. acidophilus did not change the total abundance of azoreductase (azoR) and N-acetyltransferase (NAT) associated gut microbiota and enzymes, which are involved in the metabolism of Olsalazine. Both pharmacokinetic and gut microbiota results revealed that L. acidophilus did not increase the colonic exposure of 5-ASA to improve the efficacy of combination therapy. L. acidophilus played its role in UC treatment by regulating gut microbiota composition and amino acid, phenolic acid, oligosaccharide, and peptidoglycan metabolic pathways. There was no potential medication risk of combination therapy of Olsalazine and L. acidophilus. In summary, this research provided strong evidence of medication safety and a comprehensive understanding of therapeutic advantages for combination therapy of probiotics and 5-ASA drugs from the pharmacokinetic and gut microbiota perspectives.
Collapse
Affiliation(s)
- Zhihong Li
- Center for Pharmacological Evaluation and Research of SIPI, Shanghai Institute of Pharmaceutical Industry, Hongkou, Shanghai 200437, PR China
| | - Shumei Ma
- Center for Pharmacological Evaluation and Research of SIPI, Shanghai Institute of Pharmaceutical Industry, Hongkou, Shanghai 200437, PR China; School of Pharmacy, Fudan University, Pudong, Shanghai 201203, PR China
| | - Xiaowei Wang
- Department of pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, PR China
| | - Yan Wang
- Center for Pharmacological Evaluation and Research of SIPI, Shanghai Institute of Pharmaceutical Industry, Hongkou, Shanghai 200437, PR China
| | - Renjie Yan
- Center for Pharmacological Evaluation and Research of SIPI, Shanghai Institute of Pharmaceutical Industry, Hongkou, Shanghai 200437, PR China
| | - Jiahui Wang
- Center for Pharmacological Evaluation and Research of SIPI, Shanghai Institute of Pharmaceutical Industry, Hongkou, Shanghai 200437, PR China
| | - Zhiru Xu
- Center for Pharmacological Evaluation and Research of SIPI, Shanghai Institute of Pharmaceutical Industry, Hongkou, Shanghai 200437, PR China
| | - Sheng Wang
- Center for Pharmacological Evaluation and Research of SIPI, Shanghai Institute of Pharmaceutical Industry, Hongkou, Shanghai 200437, PR China
| | - Yue Feng
- Center for Pharmacological Evaluation and Research of SIPI, Shanghai Institute of Pharmaceutical Industry, Hongkou, Shanghai 200437, PR China
| | - Juan Wang
- Center for Pharmacological Evaluation and Research of SIPI, Shanghai Institute of Pharmaceutical Industry, Hongkou, Shanghai 200437, PR China
| | - Qibing Mei
- Department of pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, PR China
| | - Ping Yang
- School of Pharmacy, Fudan University, Pudong, Shanghai 201203, PR China.
| | - Li Liu
- Center for Pharmacological Evaluation and Research of SIPI, Shanghai Institute of Pharmaceutical Industry, Hongkou, Shanghai 200437, PR China.
| |
Collapse
|
28
|
Zhou X, Liu Y, Xiong X, Chen J, Tang W, He L, Zhang Z, Yin Y, Li F. Intestinal accumulation of microbiota-produced succinate caused by loss of microRNAs leads to diarrhea in weanling piglets. Gut Microbes 2022; 14:2091369. [PMID: 35758253 PMCID: PMC9235893 DOI: 10.1080/19490976.2022.2091369] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Diarrheal disease is a common health problem with complex causality. Although diarrhea is accompanied by disturbances in microbial diversity, how gut microbes are involved in the occurrence of diarrhea remains largely unknown. Here, using a pig model of post-weaning stress-induced diarrhea, we aim to elucidate and enrich the mechanistic basis of diarrhea. We found significant alterations in fecal microbiome, their metabolites, and microRNAs levels in piglets with diarrhea. Specifically, loss of ssc-miRNA-425-5p and ssc-miRNA-423-3p, which inhibit the gene expression of fumarate reductase (frd) in Prevotella genus, caused succinate accumulation in piglets, which resulted in diarrhea. Single-cell RNA sequencing indicated impaired epithelial function and increased immune response in the colon of piglet with diarrhea. Notably, the accumulated succinate increased colonic fluid secretion by regulating transepithelial Cl-secretion in the epithelial cells. Meanwhile, succinate promoted colonic inflammatory responses by activating MyD88-dependent TLR4 signaling in the macrophages. Overall, our findings expand the mechanistic basis of diarrhea and suggest that colonic accumulation of microbiota-produced succinate caused by loss of miRNAs leads to diarrhea in weanling piglets.
Collapse
Affiliation(s)
- Xihong Zhou
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, the Chinese Academy of Sciences, Changsha, Hunan, China
| | - Yonghui Liu
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, the Chinese Academy of Sciences, Changsha, Hunan, China,Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, College of Life Sciences, Hunan Normal University, Changsha, Hunan, China
| | - Xia Xiong
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, the Chinese Academy of Sciences, Changsha, Hunan, China
| | - Jingqing Chen
- Laboratory Animal Center of the Academy of Military Medical Sciences, Beijing, China
| | - Wenjie Tang
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu, Sichuan, China
| | - Liuqin He
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, the Chinese Academy of Sciences, Changsha, Hunan, China,Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, College of Life Sciences, Hunan Normal University, Changsha, Hunan, China,CONTACT Liuqin He
| | - Zhigang Zhang
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Life Sciences, Yunnan University, Kunming, Yunnan, China
| | - Yulong Yin
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, the Chinese Academy of Sciences, Changsha, Hunan, China,Yulong Yin
| | - Fengna Li
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, the Chinese Academy of Sciences, Changsha, Hunan, China,Fengna Li No 644, Yuanda 2nd Road, Furong District, Changsha, China
| |
Collapse
|
29
|
Xue H, Song W, Wang Z, Wang Q. Ulva prolifera Polysaccharide–Manganese Alleviates Inflammation and Regulates Microbiota Composition in Dextran Sulfate Sodium-Induced Colitis Mice. Front Microbiol 2022; 13:916552. [PMID: 35722338 PMCID: PMC9205199 DOI: 10.3389/fmicb.2022.916552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Accepted: 04/20/2022] [Indexed: 11/17/2022] Open
Abstract
Manganese (Mn) deficiency exacerbates colitis symptoms, whereas diet supplemented with inorganic Mn merely maintains colon length in experimental colitis. In the present study, a new form of Mn, Ulva prolifera polysaccharide cheated-Mn (PMn) was used and its treatment effects on dextran sulfate sodium (DSS)-induced colitis were investigated. Male C57BL/6 mice were orally administrated with 3.5% DSS to induce colitis. Then, the colitis mice were treated with PBS or PMn for 7 days. The results showed that PMn administration help retrieve the body weight loss and intestinal morphology damage, and alleviate apoptosis and inflammatory responses in colitis mice. Moreover, PMn administration decreased intestinal infiltration as indicated by decreased concentration of myeloperoxidase and eosinophil peroxidase. Importantly, PMn retrieved the increased abundance of Firmicutes and the decreased abundance of Bacteroidetes caused by DSS, suggested its beneficial roles in regulating microbiota composition in mice with colon inflammation. Gut microbiota composition at the genus level in the mice administrated with PMn was similar to those in control mice, whereas they were clearly distinct from DSS-treated mice. These results support the potential therapeutic role of PMn in the treatment of intestinal colitis and microbes may play critical roles in mediating its effects.
Collapse
Affiliation(s)
- Haoran Xue
- Department of Clinical Laboratory, Qilu Hospital (Qingdao) of Shandong University, Qingdao, China
| | - Wei Song
- Ministry of Natural Resources (MNR) Key Laboratory of Marine Eco-Environmental Science and Technology, First Institute of Oceanography, Ministry of Natural Resources, Qingdao, China
| | - Zongling Wang
- Ministry of Natural Resources (MNR) Key Laboratory of Marine Eco-Environmental Science and Technology, First Institute of Oceanography, Ministry of Natural Resources, Qingdao, China
| | - Qian Wang
- Department of Clinical Laboratory, Qilu Hospital (Jinan) of Shandong University, Jinan, China
- *Correspondence: Qian Wang
| |
Collapse
|
30
|
Yan Y, Li Q, Shen L, Guo K, Zhou X. Chlorogenic acid improves glucose tolerance, lipid metabolism, inflammation and microbiota composition in diabetic db/db mice. Front Endocrinol (Lausanne) 2022; 13:1042044. [PMID: 36465648 PMCID: PMC9714618 DOI: 10.3389/fendo.2022.1042044] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 10/31/2022] [Indexed: 11/18/2022] Open
Abstract
INTRODUCTION Chronic and acute chlorogenic acid (CGA) can improve glucose tolerance (GT) and insulin sensitivity (IS). However, whether acute administration of CGA has beneficial effects on hepatic lipid metabolism and cecal microbiota composition remains unclear. METHODS In the current study, diabetic db/db mice were administered CGA or metformin, and db/m mice were used as controls to explore the effects of CGA on hepatic lipid metabolism, including fatty acid oxidation and transportation and triglyceride (TG) lipolysis and synthesis. Moreover, alterations in the inflammatory response and oxidative stress in the liver and gut microbe composition were evaluated. RESULTS The results showed that CGA decreased body weight and improved glucose tolerance and insulin resistance, and these effects were similar to those of metformin. CGA decreased hepatic lipid content by increasing the expression of CPT1a (carnitine palmitoyltransferase 1a), ACOX1 (Acyl-CoA oxidase 1), ATGL (adipose triglyceride lipase), and HSL (hormone-sensitive lipase) and decreasing that of MGAT1 (monoacylglycerol O-acyltransferase 1), DGAT1 (diacylglycerol O-acyltransferase), DGAT2, CD36, and FATP4 (fatty acid transport protein 4). Additionally, CGA restored the expression of inflammatory genes, including TNF-α (tumor necrosis factor-alpha), IL-1β (interleukin-1beta), IL-6, and IL-10, and genes encoding antioxidant enzymes, including SOD1 (superoxide dismutases 1), SOD2 (superoxide dismutases 2), and GPX1 (glutathione peroxidase 1). Furthermore, CGA improved the bacterial alpha and beta diversity in the cecum. Moreover, CGA recovered the abundance of the phylum Bacteroidetes and the genera Lactobacillus, Blautia, and Enterococcus. DISCUSSION CGA can improve the antidiabetic effects, and microbes may critically mediate these beneficial effects.
Collapse
Affiliation(s)
- Yongwang Yan
- Pharmaceutical College, Changsha Health Vocational College, Changsha, China
| | - Qing Li
- Department of Pathology, Changsha Health Vocational College, Changsha, China
| | - Ling Shen
- Pharmaceutical College, Changsha Health Vocational College, Changsha, China
| | - Kangxiao Guo
- Pharmaceutical College, Changsha Health Vocational College, Changsha, China
- National Engineering Laboratory for Rice and By-Product Deep Processing, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha, China
| | - Xu Zhou
- Department of Spleen, Stomach and Liver Diseases, Affiliated Hospital of Hunan Academy of Traditional Chinese Medicine, Changsha, China
| |
Collapse
|