1
|
Hou XW, Meng J, Chen XT, Zhao JX, Shang KM, Wei YJ, Liu R. Bacillus safensis M01 reversed the inflammatory injury of mice jejunum caused by enterotoxigenic Escherichia coli K88. Arch Microbiol 2025; 207:87. [PMID: 40087175 DOI: 10.1007/s00203-025-04287-2] [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: 01/14/2025] [Revised: 02/15/2025] [Accepted: 02/24/2025] [Indexed: 03/17/2025]
Abstract
Enterotoxigenic Escherichia coli (ETEC) is a major pathogen causing neonatal diarrhea in livestock, with antibiotics commonly used for control. However, antibiotic overuse has led to issues such as residues and bacterial resistance, underscoring the need for alternative prevention strategies. This study investigated the potential of Bacillus safensis (B. safensis) M01, isolated from healthy porcine feces in Shandong, China, to prevent ETEC infections. M01 exhibited over 80% inhibition of ETEC in vitro and was selected for further analysis. Pre-treatment of IPEC-J2 cells with M01 significantly reduced ETEC-induced cellular damage, enhanced cell viability, and inhibited bacterial adhesion. It modulated inflammatory responses by down-regulating IL-1β and TNF-α while up-regulating IL-10. Additionally, M01 promoted the expression of tight junction proteins, including Claudin-1, Occludin, and ZO-1. In the C57BL/6 mouse model, pre-feeding with M01 for 14 days improved jejunal injury caused by ETEC, as indicated by increased villus height/crypt depth ratios. Similar to in vitro findings, M01 reduced IL-1β and TNF-α expression while enhancing tight junction protein levels. These results suggest that B. safensis M01 is a promising probiotic candidate for preventing ETEC infections in livestock, offering an effective alternative to antibiotics.
Collapse
Affiliation(s)
- Xin-Wen Hou
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong Province, People's Republic of China
| | - Jinxin Meng
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong Province, People's Republic of China
| | - Xiao-Tong Chen
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong Province, People's Republic of China
| | - Ji-Xin Zhao
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong Province, People's Republic of China
| | - Kai-Meng Shang
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong Province, People's Republic of China
| | - Yong-Jie Wei
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong Province, People's Republic of China
| | - Rui Liu
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong Province, People's Republic of China.
| |
Collapse
|
2
|
Veerapagu M, Jeya K, Sankara Narayanan A. Gastrointestinal microbiome engineering in pig. HUMAN AND ANIMAL MICROBIOME ENGINEERING 2025:265-290. [DOI: 10.1016/b978-0-443-22348-8.00016-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
|
3
|
Huenchuguala S, Segura-Aguilar J. Natural Compounds That Activate the KEAP1/Nrf2 Signaling Pathway as Potential New Drugs in the Treatment of Idiopathic Parkinson's Disease. Antioxidants (Basel) 2024; 13:1125. [PMID: 39334784 PMCID: PMC11428591 DOI: 10.3390/antiox13091125] [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: 08/05/2024] [Revised: 09/03/2024] [Accepted: 09/13/2024] [Indexed: 09/30/2024] Open
Abstract
Recently, a single-neuron degeneration model has been proposed to understand the development of idiopathic Parkinson's disease based on (i) the extremely slow development of the degenerative process before the onset of motor symptoms and during the progression of the disease and (ii) the fact that it is triggered by an endogenous neurotoxin that does not have an expansive character, limiting its neurotoxic effect to single neuromelanin-containing dopaminergic neurons. It has been proposed that aminochrome is the endogenous neurotoxin that triggers the neurodegenerative process in idiopathic Parkinson's disease by triggering mitochondrial dysfunction, oxidative stress, neuroinflammation, dysfunction of both lysosomal and proteasomal protein degradation, endoplasmic reticulum stress and formation of neurotoxic alpha-synuclein oligomers. Aminochrome is an endogenous neurotoxin that is rapidly reduced by flavoenzymes and/or forms adducts with proteins, which implies that it is impossible for it to have a propagative neurotoxic effect on neighboring neurons. Interestingly, the enzymes DT-diaphorase and glutathione transferase M2-2 prevent the neurotoxic effects of aminochrome. Natural compounds present in fruits, vegetables and other plant products have been shown to activate the KEAP1/Nrf2 signaling pathway by increasing the expression of antioxidant enzymes including DT-diaphorase and glutathione transferase. This review analyzes the possibility of searching for natural compounds that increase the expression of DT-diaphorase and glutathione transferase through activation of the KEAP1/Nrf2 signaling pathway.
Collapse
Affiliation(s)
- Sandro Huenchuguala
- Escuela de Tecnología Médica, Facultad de Salud, Universidad Santo Tomás, Santiago 8370003, Chile;
| | - Juan Segura-Aguilar
- Molecular & Clinical Pharmacology, Instituto de Ciencias Biomédicas (ICBM), Faculty of Medicine, University of Chile, Santiago 8380453, Chile
| |
Collapse
|
4
|
Shi Z, Nan Y, Zhou X, Zhang W, Zhang Z, Zhang C, Duan H, Ge J, Zhao L. Molecular Mechanisms of Intestinal Protection by Levilactobacillus brevis 23017 against Salmonella typhimurium C7731-Induced Damage: Role of Nrf2. Microorganisms 2024; 12:1135. [PMID: 38930517 PMCID: PMC11205325 DOI: 10.3390/microorganisms12061135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 05/29/2024] [Accepted: 05/30/2024] [Indexed: 06/28/2024] Open
Abstract
The treatment and prevention of pathogenic diseases by lactic acid bacteria (LAB) has attracted more and more attention. As a special LAB, Levilactobacillus brevis (L. brevis) has relatively less research on its antibacterial infection in vivo, and its protective effect and mechanism still need to be fully studied. In this study, we selected L. brevis 23017, which can regulate the intestinal immunity of the host animal and resist pathogen infection, to evaluate its protective role and potential molecular mechanisms in the mouse model of S. typhimurium C7731 infection. As expected, we confirmed that L. brevis 23017 reduced the diarrhea rate and increased the daily weight gain and survival rate of the mouse model, and inhibited S. typhimurium colonization in the jejunum and liver. It also reduced the level of oxidative damage and protected the integrity of intestinal tissue by increasing the activity of intestinal antioxidant enzymes (SOD, GSH-Px and T-AOC). From the perspective of intestinal mucosal barrier injury and repair, it was confirmed that L. brevis 23017 could increase the expression levels of intestinal tight junction proteins (ZO-1 and OCLN). Our research results also show that L. brevis 23017 inhibits the inflammatory response and promotes the occurrence of cellular immunity in the body by promoting the increase in IL-10 and inhibiting IL-13 in serum and intestinal tissue. Notably, L. brevis 23017 increased total secretory immunoglobulin A (SIgA) levels in the intestine, which were closely associated with elevated levels of IL-5, IL-13, pIgR, j-chain, and IgAα-chain. In addition, L. brevis 23017 increased the expression of antioxidant proteins Nrf2, NQO1, and HO-1 associated with Nrf2 signaling to inhibit intestinal oxidative damage. This mechanism may be responsible for its protective effect against S. typhimurium-infected intestine. Our study provides new evidence and theoretical support for the analysis of the anti-bacterial infection effect and mechanism of L. brevis, which will contribute to the development of L. brevis and the treatment of pathogenic bacteria intestinal infection.
Collapse
Affiliation(s)
- Ziqi Shi
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China;
- Heilongjiang Provincial Key Laboratory of Zoonosis, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (Y.N.); (X.Z.); (W.Z.); (Z.Z.); (C.Z.); (H.D.)
| | - Yongchao Nan
- Heilongjiang Provincial Key Laboratory of Zoonosis, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (Y.N.); (X.Z.); (W.Z.); (Z.Z.); (C.Z.); (H.D.)
| | - Xinyao Zhou
- Heilongjiang Provincial Key Laboratory of Zoonosis, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (Y.N.); (X.Z.); (W.Z.); (Z.Z.); (C.Z.); (H.D.)
| | - Wenzhi Zhang
- Heilongjiang Provincial Key Laboratory of Zoonosis, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (Y.N.); (X.Z.); (W.Z.); (Z.Z.); (C.Z.); (H.D.)
| | - Zheng Zhang
- Heilongjiang Provincial Key Laboratory of Zoonosis, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (Y.N.); (X.Z.); (W.Z.); (Z.Z.); (C.Z.); (H.D.)
| | - Chuankun Zhang
- Heilongjiang Provincial Key Laboratory of Zoonosis, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (Y.N.); (X.Z.); (W.Z.); (Z.Z.); (C.Z.); (H.D.)
| | - Haoyuan Duan
- Heilongjiang Provincial Key Laboratory of Zoonosis, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (Y.N.); (X.Z.); (W.Z.); (Z.Z.); (C.Z.); (H.D.)
| | - Junwei Ge
- Heilongjiang Provincial Key Laboratory of Zoonosis, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (Y.N.); (X.Z.); (W.Z.); (Z.Z.); (C.Z.); (H.D.)
| | - Lili Zhao
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China;
| |
Collapse
|
5
|
Jin J, Liu M, Yu F, Sun MA, Wu Z. METTL3 enhances E. coli F18 resistance by targeting IKBKG/NF-κB signaling via an m 6A-YTHDF1-dependent manner in IPEC-J2 cells. Int J Biol Macromol 2024; 262:130101. [PMID: 38346619 DOI: 10.1016/j.ijbiomac.2024.130101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 01/24/2024] [Accepted: 02/08/2024] [Indexed: 02/17/2024]
Abstract
Post-weaning diarrhea caused by enterotoxigenic E. coli F18 introduces enormous losses to the porcine industry. N6-methyladenosine (m6A) is a ubiquitous epitranscriptomic biomarker that modulates host cell resistance to pathogen infection, however, its significance in E. coli F18-treated IPEC-J2 cells remains unexplored. Herein, we revealed that m6A and associated modulators strongly controlled E. coli F18 susceptibility. The data indicated an enhancement of METTL3 contents in E. coli F18-treated IPEC-J2 cells. METTL3 is known to be a major modulator of E. coli F18 adhesion within IPEC-J2 cells. As expected, METTL3 deficiency was observed to reduce m6A content at the IKBKG 5'-UTR, leading to critical suppression of YTHDF1-dependent IKBKG translation. Therefore, the activation of the NF-κB axis was observed, which enhanced IPEC-J2 resistance to E. coli F18 infection. Taken together, these findings uncover a potential mechanism underlying the m6A-mediated control of E. coli F18 susceptibility. This information may contribute to the establishment of new approaches for combating bacteria-induced diarrhea in piglets.
Collapse
Affiliation(s)
- Jian Jin
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
| | - Mengyuan Liu
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Fuying Yu
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Ming-An Sun
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
| | - Zhengchang Wu
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, China.
| |
Collapse
|
6
|
Petrariu OA, Barbu IC, Niculescu AG, Constantin M, Grigore GA, Cristian RE, Mihaescu G, Vrancianu CO. Role of probiotics in managing various human diseases, from oral pathology to cancer and gastrointestinal diseases. Front Microbiol 2024; 14:1296447. [PMID: 38249451 PMCID: PMC10797027 DOI: 10.3389/fmicb.2023.1296447] [Citation(s) in RCA: 34] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 12/18/2023] [Indexed: 01/23/2024] Open
Abstract
The imbalance of microbial composition and diversity in favor of pathogenic microorganisms combined with a loss of beneficial gut microbiota taxa results from factors such as age, diet, antimicrobial administration for different infections, other underlying medical conditions, etc. Probiotics are known for their capacity to improve health by stimulating the indigenous gut microbiota, enhancing host immunity resistance to infection, helping digestion, and carrying out various other functions. Concurrently, the metabolites produced by these microorganisms, termed postbiotics, which include compounds like bacteriocins, lactic acid, and hydrogen peroxide, contribute to inhibiting a wide range of pathogenic bacteria. This review presents an update on using probiotics in managing and treating various human diseases, including complications that may emerge during or after a COVID-19 infection.
Collapse
Affiliation(s)
- Oana-Alina Petrariu
- Microbiology-Immunology Department, Faculty of Biology, University of Bucharest, Bucharest, Romania
- The Research Institute of the University of Bucharest, Bucharest, Romania
| | - Ilda Czobor Barbu
- Microbiology-Immunology Department, Faculty of Biology, University of Bucharest, Bucharest, Romania
- The Research Institute of the University of Bucharest, Bucharest, Romania
- Academy of Romanian Scientists, Bucharest, Romania
| | - Adelina-Gabriela Niculescu
- The Research Institute of the University of Bucharest, Bucharest, Romania
- Department of Science and Engineering of Oxide Materials and Nanomaterials, Politehnica University of Bucharest, Bucharest, Romania
| | - Marian Constantin
- The Research Institute of the University of Bucharest, Bucharest, Romania
- Institute of Biology of Romanian Academy, Bucharest, Romania
| | - Georgiana Alexandra Grigore
- Microbiology-Immunology Department, Faculty of Biology, University of Bucharest, Bucharest, Romania
- The Research Institute of the University of Bucharest, Bucharest, Romania
- Academy of Romanian Scientists, Bucharest, Romania
- National Institute of Research and Development for Biological Sciences, Bucharest, Romania
| | - Roxana-Elena Cristian
- The Research Institute of the University of Bucharest, Bucharest, Romania
- National Institute of Research and Development for Biological Sciences, Bucharest, Romania
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, Bucharest, Romania
| | - Grigore Mihaescu
- Microbiology-Immunology Department, Faculty of Biology, University of Bucharest, Bucharest, Romania
| | - Corneliu Ovidiu Vrancianu
- Microbiology-Immunology Department, Faculty of Biology, University of Bucharest, Bucharest, Romania
- The Research Institute of the University of Bucharest, Bucharest, Romania
- National Institute of Research and Development for Biological Sciences, Bucharest, Romania
| |
Collapse
|
7
|
Yu C, Xu Y, Wei Y, Guo Y, Wang Y, Song P, Yu J. Gut microbiota and liver metabolomics reveal the potential mechanism of Lactobacillus rhamnosus GG modulating the liver toxicity caused by polystyrene microplastics in mice. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2024; 31:6527-6542. [PMID: 38151562 DOI: 10.1007/s11356-023-31564-8] [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: 10/13/2023] [Accepted: 12/11/2023] [Indexed: 12/29/2023]
Abstract
Microplastics (MPs) are known to cause liver toxicity as they can spread through the food chain. Most researches on their toxicity have focused on individual organs, neglecting the crucial "gut-liver axis"-a bidirectional communication pathway between the gut and liver. Probiotics have shown promise in modulating the effects of environmental pollutants. In this study, we exposed mice to Lactobacillus rhamnosus GG (LGG, 100 mg/kg b.w./d) and/or polystyrene microplastics (PS-MPs, 5 mg/kg b.w./d) for 28 d via gavage to investigate how probiotics influence live toxicity through the gut-liver axis. Our results demonstrated that PS-MPs induced liver inflammation (increased IL-6 and TNF-α) and disrupted lipid metabolism. However, when combined with LGG, these effects were alleviated. LGG also improved colon health, rectifying ciliary defects and abnormal mucus secretion caused by PS-MPs. Furthermore, LGG improved gut microbiota dysbiosis induced by PS-MPs. Metabolomics and gene expression analysis (Cyp7a1 and Cyp7b1) indicated that LGG modulated bile acid metabolism. In summary, LGG appears to protect the liver by maintaining gut homeostasis, enhancing gut barrier integrity, and reducing the liver inflammation. These findings confirm the potential of LGG to modulate liver toxicity caused by PS-MPs through the gut-liver axis, offering insights into probiotics' application for environmental pollutant detoxification.
Collapse
Affiliation(s)
- Changhao Yu
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Xuelin Road, Nanjing, 210023, People's Republic of China
| | - Yawen Xu
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Xuelin Road, Nanjing, 210023, People's Republic of China
| | - Yiping Wei
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Xuelin Road, Nanjing, 210023, People's Republic of China
| | - Yuxue Guo
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Xuelin Road, Nanjing, 210023, People's Republic of China
| | - Yi Wang
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Xuelin Road, Nanjing, 210023, People's Republic of China
| | - Ping Song
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Xuelin Road, Nanjing, 210023, People's Republic of China
| | - Jing Yu
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Xuelin Road, Nanjing, 210023, People's Republic of China.
| |
Collapse
|
8
|
Li Q, Zheng T, Ding H, Chen J, Li B, Zhang Q, Yang S, Zhang S, Guan W. Exploring the Benefits of Probiotics in Gut Inflammation and Diarrhea-From an Antioxidant Perspective. Antioxidants (Basel) 2023; 12:1342. [PMID: 37507882 PMCID: PMC10376667 DOI: 10.3390/antiox12071342] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 06/17/2023] [Accepted: 06/19/2023] [Indexed: 07/30/2023] Open
Abstract
Inflammatory bowel disease (IBD), characterized by an abnormal immune response, includes two distinct types: Crohn's disease (CD) and ulcerative colitis (UC). Extensive research has revealed that the pathogeny of IBD encompasses genetic factors, environmental factors, immune dysfunction, dysbiosis, and lifestyle choices. Furthermore, patients with IBD exhibit both local and systemic oxidative damage caused by the excessive presence of reactive oxygen species. This oxidative damage exacerbates immune response imbalances, intestinal mucosal damage, and dysbiosis in IBD patients. Meanwhile, the weaning period represents a crucial phase for pigs, during which they experience pronounced intestinal immune and inflammatory responses, leading to severe diarrhea and increased mortality rates. Pigs are highly similar to humans in terms of physiology and anatomy, making them a potential choice for simulating human IBD. Although the exact mechanism behind IBD and post-weaning diarrhea remains unclear, the oxidative damage, in its progression and pathogenesis, is well acknowledged. Besides conventional anti-inflammatory drugs, certain probiotics, particularly Lactobacillus and Bifidobacteria strains, have been found to possess antioxidant properties. These include the scavenging of reactive oxygen species, chelating metal ions to inhibit the Fenton reaction, and the regulation of host antioxidant enzymes. Consequently, numerous studies in the last two decades have committed to exploring the role of probiotics in alleviating IBD. Here, we sequentially discuss the oxidative damage in IBD and post-weaning diarrhea pathogenesis, the negative consequences of oxidative stress on IBD, the effectiveness of probiotics in IBD treatment, the application of probiotics in weaned piglets, and the potential antioxidant mechanisms of probiotics.
Collapse
Affiliation(s)
- Qihui Li
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Tenghui Zheng
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Hanting Ding
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Jiaming Chen
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Baofeng Li
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Qianzi Zhang
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Siwang Yang
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Shihai Zhang
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China
- Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou 510642, China
| | - Wutai Guan
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China
- Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou 510642, China
| |
Collapse
|
9
|
Liu X, Qiu X, Yang Y, Wang J, Wang Q, Liu J, Yang F, Liu Z, Qi R. Alteration of gut microbiome and metabolome by Clostridium butyricum can repair the intestinal dysbiosis caused by antibiotics in mice. iScience 2023; 26:106190. [PMID: 36895644 PMCID: PMC9988658 DOI: 10.1016/j.isci.2023.106190] [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: 08/15/2022] [Revised: 11/24/2022] [Accepted: 02/07/2023] [Indexed: 02/15/2023] Open
Abstract
This study evaluated the repair effects of Clostridium butyricum (CBX 2021) on the antibiotic (ABX)-induced intestinal dysbiosis in mice by the multi-omics method. Results showed that ABX eliminated more than 90% of cecal bacteria and also exerted adverse effects on the intestinal structure and overall health in mice after 10 days of the treatment. Of interest, supplementing CBX 2021 in the mice for the next 10 days colonized more butyrate-producing bacteria and accelerated butyrate production compared with the mice by natural recovery. The reconstruction of intestinal microbiota efficiently promoted the improvement of the damaged gut morphology and physical barrier in the mice. In addition, CBX 2021 significantly reduced the content of disease-related metabolites and meanwhile promoted carbohydrate digestion and absorption in mice followed the microbiome alternation. In conclusion, CBX 2021 can repair the intestinal ecology of mice damaged by the antibiotics through reconstructing gut microbiota and optimizing metabolic functions.
Collapse
Affiliation(s)
- Xin Liu
- Chongqing Academy of Animal Science, Chongqing 402460, China.,College of Animal Science and Technology, Southwest University, Chongqing 402460, China
| | - Xiaoyu Qiu
- Chongqing Academy of Animal Science, Chongqing 402460, China.,National Pig Technology Innovation Center, Chongqing 402460, China
| | - Yong Yang
- College of Life Sciences, Southwest University of Science and Technology, Mianyang 621000, China
| | - Jing Wang
- Chongqing Academy of Animal Science, Chongqing 402460, China.,National Pig Technology Innovation Center, Chongqing 402460, China
| | - Qi Wang
- Chongqing Academy of Animal Science, Chongqing 402460, China.,National Pig Technology Innovation Center, Chongqing 402460, China
| | - Jingbo Liu
- College of Life Sciences, Southwest University of Science and Technology, Mianyang 621000, China
| | - Feiyun Yang
- Chongqing Academy of Animal Science, Chongqing 402460, China.,National Pig Technology Innovation Center, Chongqing 402460, China
| | - Zuohua Liu
- Chongqing Academy of Animal Science, Chongqing 402460, China.,National Pig Technology Innovation Center, Chongqing 402460, China
| | - Renli Qi
- Chongqing Academy of Animal Science, Chongqing 402460, China.,National Pig Technology Innovation Center, Chongqing 402460, China
| |
Collapse
|
10
|
Comprehensive Analysis Revealed the Potential Roles of N6-Methyladenosine (m6A) Mediating E. coli F18 Susceptibility in IPEC-J2 Cells. Int J Mol Sci 2022; 23:ijms232113602. [DOI: 10.3390/ijms232113602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 10/20/2022] [Accepted: 11/04/2022] [Indexed: 11/11/2022] Open
Abstract
Post-weaning diarrhea caused by enterotoxigenic Escherichia coli F18 (E. coli F18) causes significant economic losses for pig producers. N6-methyladenosine (m6A) is a highly abundant epitranscriptomic marker that has been found to be involved in regulating the resistance of host cells to pathogenic infection, but its potential role in E. coli F18-exposed intestinal porcine epithelial cells (IPEC-J2) remains undetermined. Here, we demonstrated that m6A and its regulators modulate E. coli F18 susceptibility. Briefly, we revealed that the Wilms’ tumor 1-associating protein (WTAP) expressions were markedly elevated in IPEC-J2 cells upon E. coli F18 exposure. WTAP are required for the regulation of E. coli F18 adhesion in IPEC-J2 cells. Additionally, WTAP knockdown significantly suppressed m6A level at N-acetyllactosaminide beta-1,6-N-acetylglucosaminyl-transferase (GCNT2) 3′UTR, resulting in the enhancement of TH N6-methyladenosine RNA binding protein 2 (YTHDF2)-mediated GCNT2 mRNA stability. Subsequently, the altered GCNT2 expressions could inhibit the glycosphingolipid biosynthesis, thus improving resistance to E. coli F18 infection in IPEC-J2. Collectively, our analyses highlighted the mechanism behind the m6A-mediated management of E. coli F18 susceptibility, which will aid in the development of novel approaches that protect against bacterial diarrhea in piglets.
Collapse
|
11
|
Li H, Shang Z, Liu X, Qiao Y, Wang K, Qiao J. Clostridium butyricum Alleviates Enterotoxigenic Escherichia coli K88 -Induced Oxidative Damage Through Regulating the p62-Keap1-Nrf2 Signaling Pathway and Remodeling the Cecal Microbial Community. Front Immunol 2021; 12:771826. [PMID: 34899723 PMCID: PMC8660075 DOI: 10.3389/fimmu.2021.771826] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 10/28/2021] [Indexed: 12/21/2022] Open
Abstract
Clostridium butyricum (CB) can enhance antioxidant capacity and alleviate oxidative damage, but the molecular mechanism by which this occurs remains unclear. This study used enterotoxigenic Escherichia coli (ETEC) K88 as a pathogenic model, and the p62-Keap1-Nrf2 signaling pathway and intestinal microbiota as the starting point to explore the mechanism through which CB alleviates oxidative damage. After pretreatment with CB for 15 d, mice were challenged with ETEC K88 for 24 h. The results suggest that CB pretreatment can dramatically reduce crypt depth (CD) and significantly increase villus height (VH) and VH/CD in the jejunum of ETEC K88-infected mice and relieve morphological lesions of the liver and jejunum. Additionally, compared with ETEC-infected group, pretreatment with 4.4×106 CFU/mL CB can significantly reduce malondialdehyde (MDA) level and dramatically increase superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px) levels in the serum. This pretreatment can also greatly increase the mRNA expression levels of tight junction proteins and genes related to the p62-Keap1-Nrf2 signaling pathway in the liver and jejunum in ETEC K88-infected mice. Meanwhile, 16S rDNA amplicon sequencing revealed that Clostridium disporicum was significantly enriched after ETEC K88 challenge relative to the control group, while Lactobacillus was significantly enriched after 4.4×106 CFU/mL CB treatment. Furthermore, 4.4×106 CFU/mL CB pretreatment increased the short-chain fatty acid (SCFA) contents in the cecum of ETEC K88-infected mice. Moreover, we found that Lachnoclostridium, Roseburia, Lactobacillus, Terrisporobacter, Akkermansia, and Bacteroides are closely related to SCFA contents and oxidative indicators. Taken together, 4.4×106 CFU/mL CB pretreatment can alleviate ETEC K88-induced oxidative damage through activating the p62-Keap1-Nrf2 signaling pathway and remodeling the cecal microbiota community in mice.
Collapse
Affiliation(s)
- Haihua Li
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin, China
| | - Zhiyuan Shang
- Tianjin Key Laboratory of Animal and Plant Resistance, College of Life Sciences, Tianjin Normal University, Tianjin, China
| | - Xuejiao Liu
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin, China
| | - Yingying Qiao
- Faculty of Biology and Technology, Sumy National Agrarian University, Sumy, Ukraine
| | - Kewei Wang
- Tianjin Key Laboratory of Animal and Plant Resistance, College of Life Sciences, Tianjin Normal University, Tianjin, China
| | - Jiayun Qiao
- Tianjin Key Laboratory of Animal and Plant Resistance, College of Life Sciences, Tianjin Normal University, Tianjin, China
| |
Collapse
|