1
|
Jia R, Tian S, Yang Z, Lu S, Wang L, Zhang G. The mitigative role of novel aflatoxin-degrading enzymes in diverse broiler performance indicators and gut microbiota following the consumption of diets contaminated with aflatoxins. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2024; 104:7441-7453. [PMID: 38738519 DOI: 10.1002/jsfa.13564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Revised: 04/23/2024] [Accepted: 04/23/2024] [Indexed: 05/14/2024]
Abstract
BACKGROUND This study aims to explore both the toxic effects of aflatoxins (AFs) and the protective effects of degrading enzymes (DE) on broilers exposed to AFs. RESULTS The findings reveal that a diet contaminated with 69.15 μg kg-1 of aflatoxin B1 had significant adverse effects on broilers. Specifically, it led to a reduction in average daily gain, dressed yield percentage, half-eviscerated yield with giblet yield percentage, eviscerated yield percentage, as well as serum superoxide dismutase (SOD), glutathione peroxidase activity and liver SOD activity (P < 0.05). Conversely, the diet increased the feed conversion ratio, liver index, serum glutamic oxaloacetic transaminase levels and malondialdehyde levels in both serum and liver (P < 0.05). Additionally, AFs disrupted the intestinal microflora significantly (P < 0.05), altering the relative abundance of Enterococcus, Lactobacillus and Escherichia in broiler jejunum. The addition of DE to AF-contaminated feed mitigated these negative effects and reduced the residues of aflatoxin B1, aflatoxin B2 and aflatoxin M1 in the liver and duodenum (P < 0.05). We also observed that broilers fed the diet pelleted at 80 °C exhibited improved dressing percentage and water holding capacity compared to those on the 75 °C diet. CONCLUSION In summary, DE serves as an effective feed additive for mitigating AF contamination in poultry production. © 2024 Society of Chemical Industry.
Collapse
Affiliation(s)
- Ru Jia
- School of Life Science, Shanxi University, Taiyuan, China
| | - Senmiao Tian
- School of Life Science, Shanxi University, Taiyuan, China
| | - Zhaofeng Yang
- School of Life Science, Shanxi University, Taiyuan, China
| | - Simeng Lu
- School of Life Science, Shanxi University, Taiyuan, China
| | - Lan Wang
- School of Life Science, Shanxi University, Taiyuan, China
| | - Guohua Zhang
- School of Life Science, Shanxi University, Taiyuan, China
| |
Collapse
|
2
|
Kim NY, Lee SI. Lauric acid reduces apoptosis by inhibiting FOXO3a-signaling in deoxynivalenol-treated IPEC-J2 cells. JOURNAL OF ANIMAL SCIENCE AND TECHNOLOGY 2024; 66:1010-1020. [PMID: 39398305 PMCID: PMC11466732 DOI: 10.5187/jast.2023.e92] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 08/23/2023] [Accepted: 08/31/2023] [Indexed: 10/15/2024]
Abstract
Deoxynivalenol (DON) is the most common mycotoxin contaminant of food or feed worldwide and causes disease in animals. Lauric acid (LA) is a medium-chain fatty acid useful for barrier functions such as antimicrobial activity in the intestine of monogastric animals. However, the molecular mechanisms by which lauric acid exerts its effects on the deoxynivalenol-exposed small intestine have not been studied. We used an intestinal porcine epithelial cell line (IPEC-J2) as an in vitro model to explore the molecular mechanism of lauric acid in alleviating deoxynivalenol-induced damage. We found that lauric acid reversed deoxynivalenol-induced reduction in cell viability. Our quantitative real-time polymerase chain reaction results indicated that lauric acid alleviated deoxynivalenol-induced apoptosis through Annexin-V. Additionally, immunofluorescence and Western blotting showed that lauric acid attenuated deoxynivalenol-induced forkhead box O3 (FOXO3a) translocation into the nucleus. These results suggest that lauric acid attenuates forkhead box O3 translocation in the small intestine damaged by deoxynivalenol, thereby reducing apoptosis. In conclusion, this study found that lauric acid alleviates deoxynivalenol-induced damage in intestinal porcine epithelial cell line through various molecular mechanisms.
Collapse
Affiliation(s)
- Na Yeon Kim
- Department of Animal Science and
Biotechnology, Kyungpook National University, Sangju 37224,
Korea
| | - Sang In Lee
- Department of Animal Science and
Biotechnology, Kyungpook National University, Sangju 37224,
Korea
- Research Institute for Innovative Animal
Science, Kyungpook National University, Sangju 37224,
Korea
| |
Collapse
|
3
|
Maddineni G, Obulareddy SJ, Paladiya RD, Korsapati RR, Jain S, Jeanty H, Vikash F, Tummala NC, Shetty S, Ghazalgoo A, Mahapatro A, Polana V, Patel D. The role of gut microbiota augmentation in managing non-alcoholic fatty liver disease: an in-depth umbrella review of meta-analyses with grade assessment. Ann Med Surg (Lond) 2024; 86:4714-4731. [PMID: 39118769 PMCID: PMC11305784 DOI: 10.1097/ms9.0000000000002276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 06/03/2024] [Indexed: 08/10/2024] Open
Abstract
Background and aim Currently, there are no authorized medications specifically for non-alcoholic fatty liver disease (NAFLD) treatment. Studies indicate that changes in gut microbiota can disturb intestinal balance and impair the immune system and metabolism, thereby elevating the risk of developing and exacerbating NAFLD. Despite some debate, the potential benefits of microbial therapies in managing NAFLD have been shown. Methods A systematic search was undertaken to identify meta-analyses of randomized controlled trials that explored the effects of microbial therapy on the NAFLD population. The goal was to synthesize the existing evidence-based knowledge in this field. Results The results revealed that probiotics played a significant role in various aspects, including a reduction in liver stiffness (MD: -0.38, 95% CI: [-0.49, -0.26]), hepatic steatosis (OR: 4.87, 95% CI: [1.85, 12.79]), decrease in body mass index (MD: -1.46, 95% CI: [-2.43, -0.48]), diminished waist circumference (MD: -1.81, 95% CI: [-3.18, -0.43]), lowered alanine aminotransferase levels (MD: -13.40, 95% CI: [-17.02, -9.77]), decreased aspartate aminotransferase levels (MD: -13.54, 95% CI: [-17.85, -9.22]), lowered total cholesterol levels (MD: -15.38, 95% CI: [-26.49, -4.26]), decreased fasting plasma glucose levels (MD: -4.98, 95% CI: [-9.94, -0.01]), reduced fasting insulin (MD: -1.32, 95% CI: [-2.42, -0.21]), and a decline in homeostatic model assessment of insulin resistance (MD: -0.42, 95% CI: [-0.72, -0.11]) (P<0.05). Conclusion Overall, the results demonstrated that gut microbiota interventions could ameliorate a wide range of indicators including glycemic profile, dyslipidemia, anthropometric indices, and liver injury, allowing them to be considered a promising treatment strategy.
Collapse
Affiliation(s)
| | | | | | | | - Shika Jain
- MVJ Medical College and Research Hospital, Bengaluru, Karnataka, India
| | | | - Fnu Vikash
- Jacobi Medical Center, Albert Einstein College of Medicine, Bronx
| | - Nayanika C. Tummala
- Gitam Institute of Medical Sciences and Research, Visakhapatnam, Andhra Pradesh
| | | | - Arezoo Ghazalgoo
- Student Research Committee, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | | | | | - Dhruvan Patel
- Drexel University College of Medicine, Philadelphia, Pennsylvania, PA
| |
Collapse
|
4
|
Li G, Wang H, Yang J, Qiu Z, Liu Y, Wang X, Yan H, He D. The protective effects of Lactobacillus SNK-6 on growth, organ health, and intestinal function in geese exposed to low concentration Aflatoxin B1. Poult Sci 2024; 103:103904. [PMID: 38880050 PMCID: PMC11228886 DOI: 10.1016/j.psj.2024.103904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 05/22/2024] [Accepted: 05/22/2024] [Indexed: 06/18/2024] Open
Abstract
Aflatoxin B1 (AFB1) is a prevalent mycotoxin present in feed ingredients. In this study, we investigated the effects of Lactobacillus salivarius (L. salivarius) on the Landes geese exposed to AFB1. The 300 one-day-old Landes geese were randomly divided into five groups: The control group received a basic diet, while the other groups were fed a basic diet supplemented with 10 μg/kg AFB1, 10 μg/kg AFB1+ 4*108 cfu/g L. salivarius, 50 μg/kg AFB1, and 50 μg/kg AFB1 + 4*108 cfu/g L. salivarius for 63 d. Results showed that high level AFB1 exposure significantly decreased final BW and ADG, increased feed/gain ratio (F/G) and liver index (P < 0.05). L. salivarius improved levels of IL-1, IL-6, and IL-12 under low level of AFB1 exposure (P < 0.05), along with similar trends observed in serum IgA, IgG, IgM, T3, T4, TNF-ɑ, and EDT (P < 0.05). AFB1 exposure reduced jejunum villus high and villus high/crypt depth ratio, and suppressed expression of ZO-1, Occludin, and Claudin-1 mRNA, and significant improved with L. salivarius supplementation under low level AFB1 exposure (P < 0.05). AFB1 significantly increased expression levels of TLR3 and NF-kB1, with supplementation of L. salivarius showing significant improvement under low AFB1 exposure (P < 0.05). Cecal microbiota sequencing revealed that under low level AFB1 exposure, supplementation with L. salivarius increased the abundance of Bacteroidetes and Lactococcus. In summary, supplementation with 4*108 cfu/g L. salivarius under 10 μg/kg AFB1 exposure improved growth performance and immune capacity, enhanced jejunum morphology, reduced liver inflammation, altered the cecal microbial structure, and positively affected the growth and development of geese.
Collapse
Affiliation(s)
- Guangquan Li
- Institute of Agricultural Animal Husbandry and Veterinary Science, Shanghai Academy of Agricultural Sciences, Shanghai, 201100, China
| | - Huiying Wang
- Institute of Agricultural Animal Husbandry and Veterinary Science, Shanghai Academy of Agricultural Sciences, Shanghai, 201100, China
| | - Junhua Yang
- Institute for Agricultural Food Standard and Testing, Shanghai Academy of Agricultural Sciences, Shanghai 201403, China
| | - Zhi Qiu
- Institute for Agricultural Food Standard and Testing, Shanghai Academy of Agricultural Sciences, Shanghai 201403, China
| | - Yi Liu
- Institute of Agricultural Animal Husbandry and Veterinary Science, Shanghai Academy of Agricultural Sciences, Shanghai, 201100, China
| | - Xianze Wang
- Institute of Agricultural Animal Husbandry and Veterinary Science, Shanghai Academy of Agricultural Sciences, Shanghai, 201100, China
| | - Huaxiang Yan
- Institute of Agricultural Animal Husbandry and Veterinary Science, Shanghai Academy of Agricultural Sciences, Shanghai, 201100, China
| | - Daqian He
- Institute of Agricultural Animal Husbandry and Veterinary Science, Shanghai Academy of Agricultural Sciences, Shanghai, 201100, China.
| |
Collapse
|
5
|
Kim NY, Kim MO, Shin S, Kwon WS, Kim B, Lee JY, In Lee S. Effect of atractylenolide III on zearalenone-induced Snail1-mediated epithelial-mesenchymal transition in porcine intestinal epithelium. J Anim Sci Biotechnol 2024; 15:80. [PMID: 38845033 PMCID: PMC11157892 DOI: 10.1186/s40104-024-01038-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 04/18/2024] [Indexed: 06/09/2024] Open
Abstract
BACKGROUND The intestinal epithelium performs essential physiological functions, such as nutrient absorption, and acts as a barrier to prevent the entry of harmful substances. Mycotoxins are prevalent contaminants found in animal feed that exert harmful effects on the health of livestock. Zearalenone (ZEA) is produced by the Fusarium genus and induces gastrointestinal dysfunction and disrupts the health and immune system of animals. Here, we evaluated the molecular mechanisms that regulate the effects of ZEA on the porcine intestinal epithelium. RESULTS Treatment of IPEC-J2 cells with ZEA decreased the expression of E-cadherin and increased the expression of Snai1 and Vimentin, which induced Snail1-mediated epithelial-to-mesenchymal transition (EMT). In addition, ZEA induces Snail-mediated EMT through the activation of TGF-β signaling. The treatment of IPEC-J2 cells with atractylenolide III, which were exposed to ZEA, alleviated EMT. CONCLUSIONS Our findings provide insights into the molecular mechanisms of ZEA toxicity in porcine intestinal epithelial cells and ways to mitigate it.
Collapse
Affiliation(s)
- Na Yeon Kim
- Department of Animal Science and Biotechnology, Kyungpook National University, Sangju, Gyeong-sangbuk-do, 37224, Republic of Korea
| | - Myoung Ok Kim
- Department of Animal Science and Biotechnology, Kyungpook National University, Sangju, Gyeong-sangbuk-do, 37224, Republic of Korea
- Research Institute for Innovative Animal Science, Kyungpook National University, Sangju, Gyeongsangbuk-do, 37224, Republic of Korea
| | - Sangsu Shin
- Department of Animal Science and Biotechnology, Kyungpook National University, Sangju, Gyeong-sangbuk-do, 37224, Republic of Korea
- Research Institute for Innovative Animal Science, Kyungpook National University, Sangju, Gyeongsangbuk-do, 37224, Republic of Korea
| | - Woo-Sung Kwon
- Department of Animal Science and Biotechnology, Kyungpook National University, Sangju, Gyeong-sangbuk-do, 37224, Republic of Korea
- Research Institute for Innovative Animal Science, Kyungpook National University, Sangju, Gyeongsangbuk-do, 37224, Republic of Korea
| | - Bomi Kim
- National Institute for Korean Medicine Development, Gyeongsan, 38540, Republic of Korea
| | - Joon Yeop Lee
- National Institute for Korean Medicine Development, Gyeongsan, 38540, Republic of Korea
| | - Sang In Lee
- Department of Animal Science and Biotechnology, Kyungpook National University, Sangju, Gyeong-sangbuk-do, 37224, Republic of Korea.
- Research Institute for Innovative Animal Science, Kyungpook National University, Sangju, Gyeongsangbuk-do, 37224, Republic of Korea.
| |
Collapse
|
6
|
Huang L, Cao C, Lin X, Lu L, Lin X, Liu HC, Odle J, See MT, Zhang L, Wu W, Luo X, Liao X. Zinc alleviates thermal stress-induced damage to the integrity and barrier function of cultured chicken embryonic primary jejunal epithelial cells via the MAPK and PI3K/AKT/mTOR signaling pathways. Poult Sci 2024; 103:103696. [PMID: 38593549 PMCID: PMC11016803 DOI: 10.1016/j.psj.2024.103696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 03/23/2024] [Accepted: 03/25/2024] [Indexed: 04/11/2024] Open
Abstract
Zinc (Zn) could alleviate the adverse effect of high temperature (HT) on intestinal integrity and barrier function of broilers, but the underlying mechanisms remain unclear. We aimed to investigate the possible protective mechanisms of Zn on primary cultured broiler jejunal epithelial cells exposed to thermal stress (TS). In Exp.1, jejunal epithelial cells were exposed to 40℃ (normal temperature, NT) and 44℃ (HT) for 1, 2, 4, 6, or 8 h. Cells incubated for 8 h had the lowest transepithelial resistance (TEER) and the highest phenol red permeability under HT. In Exp.2, the cells were preincubated with different Zn sources (Zn sulfate as iZn and Zn proteinate with the moderate chelation strength as oZn) and Zn supplemental levels (50 and 100 µmol/L) under NT for 24 h, and then continuously incubated under HT for another 8 h. TS increased phenol red permeability, lactate dehydrogenase (LDH) activity and p-PKC/PKC level, and decreased TEER, cell proliferation, mRNA levels of claudin-1, occludin, zona occludens-1 (ZO-1), PI3K, AKT and mTOR, protein levels of claudin-1, ZO-1 and junctional adhesion molecule-A (JAM-A), and the levels of p-ERK/ERK, p-PI3K/PI3K and p-AKT/AKT. Under HT, oZn was more effective than iZn in increasing TEER, occludin, ZO-1, PI3K, and AKT mRNA levels, ZO-1 protein level, and p-AKT/AKT level; supplementation with 50 μmol Zn/L was more effective than 100 μmol Zn/L in increasing cell proliferation, JAM-A, PI3K, AKT, and PKC mRNA levels, JAM-A protein level, and the levels of p-ERK/ERK and p-PI3K/PI3K; furthermore, supplementation with 50 μmol Zn/L as oZn had the lowest LDH activity, and the highest ERK, JNK-1, and mTOR mRNA levels. Therefore, supplemental Zn, especially 50 μmol Zn/L as oZn, could alleviate the TS-induced integrity and barrier function damage of broiler jejunal epithelial cells possibly by promoting cell proliferation and tight junction protein expression via the MAPK and PI3K/AKT/mTOR signaling pathways.
Collapse
Affiliation(s)
- Liang Huang
- Mineral Nutrition Research Division, State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Chunyu Cao
- Poultry Mineral Nutrition Laboratory, College of Animal Science and Technology, Yangzhou University, Yangzhou 225000, China
| | - Xuanxu Lin
- Mineral Nutrition Research Division, State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Lin Lu
- Mineral Nutrition Research Division, State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Xi Lin
- Department of Animal Science, North Carolina State University, Raleigh, NC 27695, USA
| | - Hsiao-Ching Liu
- Department of Animal Science, North Carolina State University, Raleigh, NC 27695, USA
| | - Jack Odle
- Department of Animal Science, North Carolina State University, Raleigh, NC 27695, USA
| | - Miles Todd See
- Department of Animal Science, North Carolina State University, Raleigh, NC 27695, USA
| | - Liyang Zhang
- Mineral Nutrition Research Division, State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Wei Wu
- Poultry Mineral Nutrition Laboratory, College of Animal Science and Technology, Yangzhou University, Yangzhou 225000, China
| | - Xugang Luo
- Poultry Mineral Nutrition Laboratory, College of Animal Science and Technology, Yangzhou University, Yangzhou 225000, China
| | - Xiudong Liao
- Mineral Nutrition Research Division, State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China.
| |
Collapse
|
7
|
Ding S, Li K, Han X, Lin W, Qin Y, Cao R, Ren Y. Long-term use of etomidate disrupts the intestinal homeostasis and nervous system in mice. Toxicology 2024; 504:153802. [PMID: 38604439 DOI: 10.1016/j.tox.2024.153802] [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: 02/09/2024] [Revised: 04/07/2024] [Accepted: 04/09/2024] [Indexed: 04/13/2024]
Abstract
Etomidate (ETO) is used as an anesthetic in surgery, but it is being abused in some populations. The damage caused by long-term intake of ETO to intestinal and brain functions is not yet clear, and it remains to be determined whether the drug affects the central nervous system through the gut-brain axis. This study aimed to investigate the neurotoxic and gastrointestinal effects of ETO at doses of 1 mg/kg and 3 mg/kg in mice over 14 consecutive days. The results showed that long-term injection of ETO led to drug resistance in mice, affecting their innate preference for darkness and possibly inducing dependence on ETO. The levels of 5-hydroxytryptamine in the brain, serum, and colon decreased by 37%, 51%, and 42% respectively, while the levels of γ-aminobutyric acid reduced by 38%, 52%, and 41% respectively. H&E staining revealed that ETO reduced goblet cells in the colon and damaged the intestinal barrier. The expression of tight junction-related genes Claudin4 and ZO-1 was downregulated. The intestinal flora changed, the abundance of Akkermansia and Lactobacillus decreased by 33% and 14%, respectively, while Klebsiella increased by 18%. TUNEL results showed that high-dose ETO increased apoptotic cells in the brain. The expression of Claudin1 in the brain was downregulated. Untargeted metabolomics analysis of the colon and brain indicated that ETO caused abnormalities in glycerophospholipid metabolism. Abnormal lipid metabolism might lead to the production or accumulation of lipotoxic metabolites, causing central nervous system diseases. ETO induced changes in the intestinal flora and metabolism, further affecting the central nervous system through the gut-brain axis. The study unveiled the detrimental effects on the brain and gastrointestinal system resulting from long-term intake of ETO, which holds significant implications for comprehending the adverse impact of ETO abuse on human health.
Collapse
Affiliation(s)
- Siming Ding
- School of Environment and Energy, South China University of Technology, Guangzhou 510006, PR China
| | - Kan Li
- National Anti-Drug Laboratory Guangdong Regional Center, Guangzhou 510230, PR China; Anti-Drug Technology Center of Guangdong Province, Guangzhou 510230, PR China
| | - Xing Han
- National Anti-Drug Laboratory Guangdong Regional Center, Guangzhou 510230, PR China; Anti-Drug Technology Center of Guangdong Province, Guangzhou 510230, PR China
| | - Wenting Lin
- School of Environment and Energy, South China University of Technology, Guangzhou 510006, PR China
| | - Yingjun Qin
- School of Environment and Energy, South China University of Technology, Guangzhou 510006, PR China
| | - Renjuan Cao
- School of Environment and Energy, South China University of Technology, Guangzhou 510006, PR China
| | - Yuan Ren
- School of Environment and Energy, South China University of Technology, Guangzhou 510006, PR China; The Key Lab of Pollution Control and Ecosystem Restoration in Industry Clusters, Ministry of Education, Guangzhou 510006, PR China; The Key Laboratory of Environmental Protection and Eco-Remediation of Guangdong Regular Higher Education Institutions, Guangzhou 510006, PR China.
| |
Collapse
|
8
|
Jannah MW, Handayani F, Lukiswanto BS, Arif MAA, Suwarno S, Purnobasuki H, Sugihartuti R, Utama S, Darodjah S, Lestari TD, Lamid M, Jang G, Safitri E. Investigation of a multicomponent mycotoxin detoxifying agent for aflatoxin B1 and ochratoxin A-induced blood profile in broiler chickens. Vet World 2024; 17:1044-1051. [PMID: 38911087 PMCID: PMC11188902 DOI: 10.14202/vetworld.2024.1044-1051] [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: 12/31/2023] [Accepted: 04/18/2024] [Indexed: 06/25/2024] Open
Abstract
Background and Aim Mycotoxins such as aflatoxin B1 and ochratoxin A (OTA) are secondary metabolites in molds that grow in raw materials or commercial feed. This interaction has a synergistic effect on mortality, body weight, feed intake, embryo abnormalities, egg production, and lymphoid organ atrophy. This study was conducted to determine the effect of a mycotoxin detoxifier on the blood profile of broilers that were given feed contaminated with mycotoxin, such as the number of heterophils, lymphocytes, monocytes, mean corpuscular hemoglobin (MCH), and MCH concentration (MCHC). Materials and Methods A total of 20 day-old chicks (DOC) of Cobb broilers were given four treatments with five replicates. The number of chickens used in this research was determined using statistical calculations, and the data obtained was homogeneous so that the population was represented. Treatments included negative control with basal feed (C-), positive control with mycotoxins contamination (C+), treatment 1: Mycotoxins contamination and mycotoxin detoxification 1.1 g/kg (T1), and treatment 2: Mycotoxins contamination and mycotoxin detoxification 1.6 g/kg (T2). Mycotoxin contamination comprised 0.1 mg/kg aflatoxin B1 and 0.1 mg/kg OTA. The treatment period for chickens was 28 days, from 8 to 35 days. A battery cage was used in this study. Chickens were kept in a closed, ventilated room and the room temperature (27°C) was monitored during the treatment period. Results Based on the results of statistical data processing, a significant difference (p < 0.05) was observed between chickens fed mycotoxin-contaminated feed (C+) and chickens not fed mycotoxin-contaminated feed (C-) and chickens given 1.6 g/kg mycotoxin detoxification (T2). Mycotoxin detoxification at a dose of 1.6 g/kg had a significant (p < 0.05) effect on the heterophil, lymphocyte, and heterophil lymphocyte ratio, leukocyte, erythrocyte, and hemoglobin levels of the blood broiler in this experiment. On other parameters such as monocytes, MCH, and MCHC, treatment 2 at dose 1.6 g/kg was the best treatment, although there was no significant effect with C- and T1. Conclusion The administration of mycotoxin detoxifiers at a dose of 1.6 g/kg increased the number of heterophils and the ratio of heterophil lymphocytes, leukocytes, erythrocytes, and hemoglobin in broilers fed mycotoxin-contaminated feed.
Collapse
Affiliation(s)
- Mutmainah Wardatul Jannah
- Department of Veterinary Medicine, Student of Veterinary Medicine Faculty, Universitas Airlangga, Surabaya, East Java, Indonesia
| | - Fitri Handayani
- Department of Veterinary Medicine, Student of Veterinary Medicine Faculty, Universitas Airlangga, Surabaya, East Java, Indonesia
| | - Bambang Sektiari Lukiswanto
- Veterinary Clinic Division of Veterinary Medicine Faculty, Universitas Airlangga, Surabaya, East Java, Indonesia
| | - Mohammad Anam Al Arif
- Veterinary Animal Husbandry Division of Veterinary Medicine Faculty, Universitas Airlangga, Surabaya, East Java, Indonesia
| | - Suwarno Suwarno
- Veterinary Microbiology Division of Veterinary Medicine Faculty, Universitas Airlangga, Surabaya, East Java, Indonesia
| | - Hery Purnobasuki
- Department of Biology, Faculty of Science and Technology, Universitas Airlangga, Surabaya, East Java, Indonesia
| | - Rahmi Sugihartuti
- Basic Veterinary Medicine Division of Veterinary Medicine Faculty, Universitas Airlangga, Surabaya, East Java, Indonesia
| | - Suzanita Utama
- Veterinary Reproduction Division of Veterinary Medicine Faculty, Universitas Airlangga, Surabaya, East Java, Indonesia
| | - Siti Darodjah
- Department of Animal Production, Animal Husbandry Faculty, Universitas Padjadjaran, West Java Indonesia
| | - Tita Damayanti Lestari
- Veterinary Reproduction Division of Veterinary Medicine Faculty, Universitas Airlangga, Surabaya, East Java, Indonesia
| | - Mirni Lamid
- Veterinary Animal Husbandry Division of Veterinary Medicine Faculty, Universitas Airlangga, Surabaya, East Java, Indonesia
| | - Goo Jang
- Department of Theriogenology, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Erma Safitri
- Veterinary Reproduction Division of Veterinary Medicine Faculty, Universitas Airlangga, Surabaya, East Java, Indonesia
| |
Collapse
|
9
|
Song Y, Sun M, Ma F, Xu D, Mu G, Jiao Y, Yu P, Tuo Y. Lactiplantibacillus plantarum DLPT4 Protects Against Cyclophosphamide-Induced Immunosuppression in Mice by Regulating Immune Response and Intestinal Flora. Probiotics Antimicrob Proteins 2024; 16:321-333. [PMID: 36715883 DOI: 10.1007/s12602-022-10015-9] [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] [Accepted: 11/21/2022] [Indexed: 01/31/2023]
Abstract
In this study, the strain Lactiplantibacillus plantarum DLPT4 was investigated for the immunostimulatory activity in cyclophosphamide (CTX)-induced immunosuppressed BALB/c mice. L. plantarum DLPT4 was administered to BALB/c mice by oral gavage for 30 days, and CTX was injected intraperitoneally from the 25th to the 27th days. Intraperitoneal injection of CTX caused damage to the thymic cortex and intestines, and the immune dysfunction of the BALB/c mice. L. plantarum DLPT4 oral administration exerted immunoregulating effects evidenced by increasing serum immunoglobulin (IgA, IgG, and IgM) levels and reducing the genes expression of pro-inflammatory factors (IL-6, IL-1β, and TNF-α) of the CTX-induced immunosuppressed mice. The results of the metagenome-sequencing analysis showed that oral administration of L. plantarum DLPT4 could regulate the intestinal microbial community of the immunosuppressed mice by changing the ratio of Lactiplantibacillus and Bifidobacterium. Meanwhile, the abundance of carbohydrate enzyme (CAZyme), immune diseases metabolic pathways, and AP-1/MAPK signaling pathways were enriched in the mice administrated with L. plantarum DLPT4. In conclusion, oral administration of L. plantarum DLPT4 ameliorated symptoms of CTX-induced immunosuppressed mice by regulating gut microbiota, influencing the abundance of carbohydrate esterase in the intestinal flora, and enhancing immune metabolic activity. L. plantarum DLPT4 could be a potential probiotic to regulate the immune response.
Collapse
Affiliation(s)
- Yinglong Song
- School of Food Science and Technology, Dalian Polytechnic University, Dalian, 116034, People's Republic of China
- Dalian Probiotics Function Research Key Laboratory, Dalian Polytechnic University, Dalian, 116034, People's Republic of China
| | - Mengying Sun
- School of Food Science and Technology, Dalian Polytechnic University, Dalian, 116034, People's Republic of China
- Dalian Probiotics Function Research Key Laboratory, Dalian Polytechnic University, Dalian, 116034, People's Republic of China
| | - Fenglian Ma
- School of Food Science and Technology, Dalian Polytechnic University, Dalian, 116034, People's Republic of China
- Dalian Probiotics Function Research Key Laboratory, Dalian Polytechnic University, Dalian, 116034, People's Republic of China
| | - Dongxue Xu
- School of Food Science and Technology, Dalian Polytechnic University, Dalian, 116034, People's Republic of China
- Dalian Probiotics Function Research Key Laboratory, Dalian Polytechnic University, Dalian, 116034, People's Republic of China
| | - Guangqing Mu
- School of Food Science and Technology, Dalian Polytechnic University, Dalian, 116034, People's Republic of China
- Dalian Probiotics Function Research Key Laboratory, Dalian Polytechnic University, Dalian, 116034, People's Republic of China
| | - Yang Jiao
- College of Life Science and Engineering of Hexi University, Zhangye, 734000, People's Republic of China
| | - Ping Yu
- High Change (Shenyang) Child-Food Products Co, Ltd, Shenyang, 110011, People's Republic of China
| | - Yanfeng Tuo
- School of Food Science and Technology, Dalian Polytechnic University, Dalian, 116034, People's Republic of China.
- Dalian Probiotics Function Research Key Laboratory, Dalian Polytechnic University, Dalian, 116034, People's Republic of China.
| |
Collapse
|
10
|
Wang Y, Wang X, Zhu YC, Wang D, Lv L, Chen L, Jin Y. Co-exposure ochratoxin A and triadimefon influenced the hepatic glucolipid metabolism and intestinal micro-environment in mice. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 912:169339. [PMID: 38103602 DOI: 10.1016/j.scitotenv.2023.169339] [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: 10/12/2023] [Revised: 12/11/2023] [Accepted: 12/11/2023] [Indexed: 12/19/2023]
Abstract
Ochratoxin A (OTA) is a mycotoxin, and triadimefon (TDF) is a triazole fungicide. These compounds are prevalent in the environment, and their residues have been detected in crops. However, the precise health risks associated with mycotoxins and fungicides are not fully elucidated. In this work, five-week-old mice were gavage with OTA (0.3 and 1.5 mg/kg/day), TDF (10 and 50 mg/kg/day), and OTA + TDF (0.3 + 10 and 1.5 + 50 mg/kg/day) for 28 days. Exposure to OTA, TDF, and OTA + TDF led to significant alterations in liver total cholesterol (TC), triglyceride (TG), and glucose (GLU) levels, as well as in genes associated with glycolipid metabolism in mice. Reduced acylcarnitine levels in serum indicated that OTA, TDF, and co-exposure inhibited fatty acid (FA) β-oxidation. Furthermore, OTA and TDF disrupted the integrality of the gut barrier function and altered the structure of the intestinal microbiota. These findings suggested that OTA, TDF, and their co-exposure might disrupt the intestinal barrier, alter the structure of the microbiota, and subsequently inhibit FA β-oxidation, indicating the interference of OTA and TDF with glycolipid-related intestinal barrier dysfunction. Moreover, our data revealed a toxic additive effect between OTA and TDF, providing a foundation for assessing the combined toxicity risk of mycotoxins and fungicides.
Collapse
Affiliation(s)
- Yanhua Wang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Institute of Quality and Standard for Agro-products, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, Zhejiang, PR China
| | - Xiaofang Wang
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, Zhejiang, PR China
| | - Yu-Cheng Zhu
- United States Department of Agriculture, Agricultural Research Service (USDA-ARS), 141 Experiment Station Road, Stoneville, MS 38776, USA
| | - Dou Wang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Institute of Quality and Standard for Agro-products, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, Zhejiang, PR China
| | - Lu Lv
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Institute of Quality and Standard for Agro-products, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, Zhejiang, PR China
| | - Liezhong Chen
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Institute of Quality and Standard for Agro-products, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, Zhejiang, PR China.
| | - Yuanxiang Jin
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, Zhejiang, PR China.
| |
Collapse
|
11
|
Zhang DW, Lu JL, Dong BY, Fang MY, Xiong X, Qin XJ, Fan XM. Gut microbiota and its metabolic products in acute respiratory distress syndrome. Front Immunol 2024; 15:1330021. [PMID: 38433840 PMCID: PMC10904571 DOI: 10.3389/fimmu.2024.1330021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 01/30/2024] [Indexed: 03/05/2024] Open
Abstract
The prevalence rate of acute respiratory distress syndrome (ARDS) is estimated at approximately 10% in critically ill patients worldwide, with the mortality rate ranging from 17% to 39%. Currently, ARDS mortality is usually higher in patients with COVID-19, giving another challenge for ARDS treatment. However, the treatment efficacy for ARDS is far from satisfactory. The relationship between the gut microbiota and ARDS has been substantiated by relevant scientific studies. ARDS not only changes the distribution of gut microbiota, but also influences intestinal mucosal barrier through the alteration of gut microbiota. The modulation of gut microbiota can impact the onset and progression of ARDS by triggering dysfunctions in inflammatory response and immune cells, oxidative stress, cell apoptosis, autophagy, pyroptosis, and ferroptosis mechanisms. Meanwhile, ARDS may also influence the distribution of metabolic products of gut microbiota. In this review, we focus on the impact of ARDS on gut microbiota and how the alteration of gut microbiota further influences the immune function, cellular functions and related signaling pathways during ARDS. The roles of gut microbiota-derived metabolites in the development and occurrence of ARDS are also discussed.
Collapse
Affiliation(s)
- Dong-Wei Zhang
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Inflammation & Allergic Diseases Research Unit, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Department of Respiratory and Critical Care Medicine, Liuzhou People’s Hospital, Guangxi Medical University, Liuzhou, Guangxi, China
- Key Laboratory of Diagnosis, Treatment and Research of Asthma and Chronic Obstructive Pulmonary Disease, Liuzhou, Guangxi, China
| | - Jia-Li Lu
- Department of Respiratory and Critical Care Medicine, Liuzhou People’s Hospital, Guangxi Medical University, Liuzhou, Guangxi, China
- Key Laboratory of Diagnosis, Treatment and Research of Asthma and Chronic Obstructive Pulmonary Disease, Liuzhou, Guangxi, China
| | - Bi-Ying Dong
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Inflammation & Allergic Diseases Research Unit, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Department of Respiratory and Critical Care Medicine, Liuzhou People’s Hospital, Guangxi Medical University, Liuzhou, Guangxi, China
- Key Laboratory of Diagnosis, Treatment and Research of Asthma and Chronic Obstructive Pulmonary Disease, Liuzhou, Guangxi, China
| | - Meng-Ying Fang
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Inflammation & Allergic Diseases Research Unit, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Xia Xiong
- Department of Dermatology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Xue-Jun Qin
- Department of Respiratory and Critical Care Medicine, Liuzhou People’s Hospital, Guangxi Medical University, Liuzhou, Guangxi, China
- Key Laboratory of Diagnosis, Treatment and Research of Asthma and Chronic Obstructive Pulmonary Disease, Liuzhou, Guangxi, China
| | - Xian-Ming Fan
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Inflammation & Allergic Diseases Research Unit, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
12
|
Yang L, Liao W, Dong J, Chen X, Huang L, Yang W, Jiang S. Zearalenone Promotes Uterine Hypertrophy through AMPK/mTOR Mediated Autophagy. Toxins (Basel) 2024; 16:73. [PMID: 38393151 PMCID: PMC10892946 DOI: 10.3390/toxins16020073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Revised: 01/16/2024] [Accepted: 01/17/2024] [Indexed: 02/25/2024] Open
Abstract
Zearalenone (ZEN), a non-steroidal Fusarium graminearum with an estrogen effect, can cause damage to the gastrointestinal tract, immune organs, liver, and reproductive system. Further analysis of the mechanism of ZEN has become an important scientific issue. We have established in vivo and in vitro models of ZEN intervention, used AMPK/mTOR as a targeted pathway for ZEN reproductive toxicity, and explored the molecular mechanism by which ZEN may induce uterine hypertrophy in weaned piglets. Our study strongly suggested that ZEN can activate the phosphorylation of AMPK in uterine endometrial epithelium cells, affect the phosphorylation level of mTOR through TSC2 and Rheb, induce autophagy, upregulate the expression of proliferative genes PCNA and BCL2, downregulate the expression of apoptotic gene BAX, promote uterine endometrial epithelium cells proliferation, and ultimately lead to thickening of the endometrial and myometrium, increased density of uterine glands, and induce uterine hypertrophy.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Shuzhen Jiang
- Key Laboratory of Efficient Utilization of Non-Grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, College of Animal Sciences and Veterinary Medicine, Shandong Agricultural University, Tai’an 271018, China; (L.Y.); (W.L.); (J.D.); (X.C.); (L.H.); (W.Y.)
| |
Collapse
|
13
|
Liu Y, Liu G, Fang J. Progress on the mechanisms of Lactobacillus plantarum to improve intestinal barrier function in ulcerative colitis. J Nutr Biochem 2024; 124:109505. [PMID: 37890709 DOI: 10.1016/j.jnutbio.2023.109505] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 10/22/2023] [Accepted: 10/24/2023] [Indexed: 10/29/2023]
Abstract
Ulcerative colitis (UC) is a chronic, non-specific inflammatory sickness of the intestinal tract, chiefly implicating the rectum and colon, which is characterized by chronic or subacute diarrhea, mucopurulent stools, and abdominal pain. The pathogeny of UC is still uncertain, and it is thought that multiple factors interact to cause the disease, such as environment, genetics, gut microbes, and immunity. Injuring the intestinal barrier is one of the most significant features of UC and includes mechanical, chemical, immune, and biological barriers. Plenty of research has shown that probiotics, as profitable bacteria in the gut, can play a prominent role in the treatment of UC by improving gut barrier function and modulating gut immunity. Lactobacillus plantarum (L. plantarum), a common probiotic, has made outstanding contributions to food and medicine, and many studies in recent years have shown that L. plantarum has great preventive and therapeutic effects on ulcerative colitis and restores the intestinal barrier. This paper reviews the mechanisms of L. plantarum for improving the intestinal barrier function of UC organisms, mainly including regulating the immune response, inhibiting oxidative stress, raising the expression of tight junction (TJ) proteins, promoting the formation of mucin, improving the composition of gut flora, and raising the levels of short-chain fatty acids (SCFAs), which offers some help for the clinical therapy of UC.
Collapse
Affiliation(s)
- Yihui Liu
- College of Bioscience and Biotechnology, Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, Hunan Agricultural University, 1 Nongda Road, Changsha, Hunan 410128, China
| | - Gang Liu
- College of Bioscience and Biotechnology, Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, Hunan Agricultural University, 1 Nongda Road, Changsha, Hunan 410128, China.
| | - Jun Fang
- College of Bioscience and Biotechnology, Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, Hunan Agricultural University, 1 Nongda Road, Changsha, Hunan 410128, China.
| |
Collapse
|
14
|
Hu P, Yuan M, Guo B, Lin J, Yan S, Huang H, Chen JL, Wang S, Ma Y. Citric Acid Promotes Immune Function by Modulating the Intestinal Barrier. Int J Mol Sci 2024; 25:1239. [PMID: 38279237 PMCID: PMC10817003 DOI: 10.3390/ijms25021239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 01/09/2024] [Accepted: 01/16/2024] [Indexed: 01/28/2024] Open
Abstract
Amidst increasing concern about antibiotic resistance resulting from the overuse of antibiotics, there is a growing interest in exploring alternative agents. One such agent is citric acid, an organic compound commonly used for various applications. Our research findings indicate that the inclusion of citric acid can have several beneficial effects on the tight junctions found in the mouse intestine. Firstly, the study suggests that citric acid may contribute to weight gain by stimulating the growth of intestinal epithelial cells (IE-6). Citric acid enhances the small intestinal villus-crypt ratio in mice, thereby promoting intestinal structural morphology. Additionally, citric acid has been found to increase the population of beneficial intestinal microorganisms, including Bifidobacterium and Lactobacillus. It also promotes the expression of important protein genes such as occludin, ZO-1, and claudin-1, which play crucial roles in maintaining the integrity of the tight junction barrier in the intestines. Furthermore, in infected IEC-6 cells with H9N2 avian influenza virus, citric acid augmented the expression of genes closely associated with the influenza virus infection. Moreover, it reduces the inflammatory response caused by the viral infection and thwarted influenza virus replication. These findings suggest that citric acid fortifies the intestinal tight junction barrier, inhibits the replication of influenza viruses targeting the intestinal tract, and boosts intestinal immune function.
Collapse
Affiliation(s)
- Pengcheng Hu
- Joint Laboratory of Animal Pathogen Prevention and Control of Fujian-Nepal, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China; (P.H.); (M.Y.); (S.Y.); (J.-L.C.); (S.W.)
| | - Meng Yuan
- Joint Laboratory of Animal Pathogen Prevention and Control of Fujian-Nepal, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China; (P.H.); (M.Y.); (S.Y.); (J.-L.C.); (S.W.)
| | - Bolun Guo
- Key Laboratory of Fujian-Taiwan Animal Pathogen Biology, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China; (B.G.); (J.L.); (H.H.)
| | - Jiaqi Lin
- Key Laboratory of Fujian-Taiwan Animal Pathogen Biology, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China; (B.G.); (J.L.); (H.H.)
| | - Shihong Yan
- Joint Laboratory of Animal Pathogen Prevention and Control of Fujian-Nepal, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China; (P.H.); (M.Y.); (S.Y.); (J.-L.C.); (S.W.)
- Key Laboratory of Fujian-Taiwan Animal Pathogen Biology, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China; (B.G.); (J.L.); (H.H.)
| | - Huiqing Huang
- Key Laboratory of Fujian-Taiwan Animal Pathogen Biology, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China; (B.G.); (J.L.); (H.H.)
| | - Ji-Long Chen
- Joint Laboratory of Animal Pathogen Prevention and Control of Fujian-Nepal, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China; (P.H.); (M.Y.); (S.Y.); (J.-L.C.); (S.W.)
- Key Laboratory of Fujian-Taiwan Animal Pathogen Biology, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China; (B.G.); (J.L.); (H.H.)
| | - Song Wang
- Joint Laboratory of Animal Pathogen Prevention and Control of Fujian-Nepal, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China; (P.H.); (M.Y.); (S.Y.); (J.-L.C.); (S.W.)
- Key Laboratory of Fujian-Taiwan Animal Pathogen Biology, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China; (B.G.); (J.L.); (H.H.)
| | - Yanmei Ma
- Joint Laboratory of Animal Pathogen Prevention and Control of Fujian-Nepal, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China; (P.H.); (M.Y.); (S.Y.); (J.-L.C.); (S.W.)
- Key Laboratory of Fujian-Taiwan Animal Pathogen Biology, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China; (B.G.); (J.L.); (H.H.)
| |
Collapse
|
15
|
Hou S, Ma J, Cheng Y, Wang Z, Wang G, Jia A, Wang H, Sun J, Yan Y. DON induced DNA damage triggers absence of p53-mediated G2 arrest and apoptosis in IPEC-1 cells. Toxicology 2024; 501:153707. [PMID: 38104654 DOI: 10.1016/j.tox.2023.153707] [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: 10/10/2023] [Revised: 12/11/2023] [Accepted: 12/12/2023] [Indexed: 12/19/2023]
Abstract
Deoxynivalenol (DON) stands among the prevalent mycotoxins, and usually contaminates cereal foods and animal feed, leading to human and animal clinical poisoning symptoms such as abdominal pain, diarrhea, and vomiting. To date, the mechanism of toxicity of DON in different mammalian cells is not fully elucidated. In this study, we explored the detrimental impacts of DON on porcine intestinal epithelial cells (IPEC-1), serving as a representative model for porcine intestinal epithelial cells. After treating cells with DON for 24 h, DON can significantly inhibit the activity of cells, induce the production of reactive oxygen species (ROS), significantly reduce the content of glutathione and the activity of catalase, and increase the activity of superoxide dismutase and malondialdehyde, leading to an imbalance in intracellular redox status. In addition, DON can induce DNA double-strand breaks, and decrease mitochondrial membrane potential. Furthermore, DON can promote the release of Cyt C through changes in mitochondrial permeability through inhibit the expression of B-cell lymphoma 2 (Bcl-2) proteins, leading to apoptosis through the mitochondrial pathway. On the other hand, we found that DON can cause IPEC-1 cells G2 phase cycle arrest. Different with our pervious study, DON induces cell cycle arrest in the G2 phase only by activating the ATM-Chk2-Cdc 25 C pathway, but cannot regulate the cell cycle arrest via the ATM-p53 pathway. These results indicate that DON can induce the same toxic phenotype in different cells, but its toxic mechanism is different. All these provide a rationale for revealing DON induced cytotoxicity and intestinal diseases.
Collapse
Affiliation(s)
- Silu Hou
- Shanghai Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, No. 800 Dongchuan Road, Shanghai 200240, China; State Key Laboratory of Food Nutrition and Safety, College of Food Science and Engineering, Demonstration Center of Food Quality and Safety Testing Technology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Jingjiao Ma
- Shanghai Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, No. 800 Dongchuan Road, Shanghai 200240, China
| | - Yuqiang Cheng
- Shanghai Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, No. 800 Dongchuan Road, Shanghai 200240, China
| | - Zhaofei Wang
- Shanghai Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, No. 800 Dongchuan Road, Shanghai 200240, China
| | - Guiping Wang
- Guangdong Haid Group Co. Limited., Guangzhou, Guangdong 511400, China
| | - Aiqing Jia
- Guangdong Haid Group Co. Limited., Guangzhou, Guangdong 511400, China
| | - Hengan Wang
- Shanghai Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, No. 800 Dongchuan Road, Shanghai 200240, China
| | - Jianhe Sun
- Shanghai Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, No. 800 Dongchuan Road, Shanghai 200240, China
| | - Yaxian Yan
- Shanghai Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, No. 800 Dongchuan Road, Shanghai 200240, China.
| |
Collapse
|
16
|
Liu B, Deng Y, Duan Z, Chu C, Wang X, Yang C, Li J, Ding W. Neutrophil extracellular traps promote intestinal barrier dysfunction by regulating macrophage polarization during trauma/hemorrhagic shock via the TGF-β signaling pathway. Cell Signal 2024; 113:110941. [PMID: 37890686 DOI: 10.1016/j.cellsig.2023.110941] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 10/01/2023] [Accepted: 10/24/2023] [Indexed: 10/29/2023]
Abstract
The mechanism by which neutrophil extracellular traps (NETs) may cause intestinal barrier dysfunction in response to trauma/hemorrhagic shock (T/HS) remains unclear. In this study, the roles and mechanisms of NETs in macrophage polarization were examined to determine whether this process plays a role in tissue damage associated with T/HS. Rat models of T/HS and macrophage polarization were developed and the levels of NETs formation in the intestinal tissue of T/HS rats were assessed. NET formation was inhibited in models of T/HS to examine the effect on intestinal inflammation and barrier injury. The proportions of pro-inflammatory and anti-inflammatory macrophages in the damaged intestinal tissues were measured. Finally, high-throughput sequencing was performed to investigate the underlying mechanisms involved in this process. The study revealed that the level of NETs formation was increased and that inhibition of NETs formation alleviated the intestinal inflammation and barrier injury. Moreover, the number of pro-inflammatory macrophages increased and the number of anti-inflammatory macrophages decreased. RNA sequencing analysis indicated that NETs formation decreased the expression of transforming growth factor-beta receptor 2 (TGFBR2), bioinformatic analyses revealed that TGFBR2 was significantly enriched in the transforming growth factor-beta (TGF-β) signaling pathway. Verification experiments showed that NETs impeded macrophage differentiation into the anti-inflammatory/M2 phenotype and inhibited TGFBR2 and TGF-β expression in macrophages. However, treatment with DNase I and overexpression of TGFBR2, and inhibition of TGF-β promoted and prevented this process, respectively. NETs may regulate the macrophage polarization process by promoting intestinal barrier dysfunction in T/HS rats through the TGFBR2-mediated TGF-β signaling pathway.
Collapse
Affiliation(s)
- Baochen Liu
- Department of Trauma and Acute Care Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yunxuan Deng
- Department of Trauma and Acute Care Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Zehua Duan
- Department of Trauma and Acute Care Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Chengnan Chu
- Department of Trauma and Acute Care Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xingyu Wang
- Department of Trauma and Acute Care Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Chao Yang
- Department of Trauma and Acute Care Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Jieshou Li
- Department of Trauma and Acute Care Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| | - Weiwei Ding
- Department of Trauma and Acute Care Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| |
Collapse
|
17
|
Liu Y, Li G, Lu F, Guo Z, Cai S, Huo T. Excess iron intake induced liver injury: The role of gut-liver axis and therapeutic potential. Biomed Pharmacother 2023; 168:115728. [PMID: 37864900 DOI: 10.1016/j.biopha.2023.115728] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 10/10/2023] [Accepted: 10/13/2023] [Indexed: 10/23/2023] Open
Abstract
Excessive iron intake is detrimental to human health, especially to the liver, which is the main organ for iron storage. Excessive iron intake can lead to liver injury. The gut-liver axis (GLA) refers to the bidirectional relationship between the gut and its microbiota and the liver, which is a combination of signals generated by dietary, genetic and environmental factors. Excessive iron intake disrupts the GLA at multiple interconnected levels, including the gut microbiota, gut barrier function, and the liver's innate immune system. Excessive iron intake induces gut microbiota dysbiosis, destroys gut barriers, promotes liver exposure to gut microbiota and its derived metabolites, and increases the pro-inflammatory environment of the liver. There is increasing evidence that excess iron intake alters the levels of gut microbiota-derived metabolites such as secondary bile acids (BAs), short-chain fatty acids, indoles, and trimethylamine N-oxide, which play an important role in maintaining homeostasis of the GLA. In addition to iron chelators, antioxidants, and anti-inflammatory agents currently used in iron overload therapy, gut barrier intervention may be a potential target for iron overload therapy. In this paper, we review the relationship between excess iron intake and chronic liver diseases, the regulation of iron homeostasis by the GLA, and focus on the effects of excess iron intake on the GLA. It has been suggested that probiotics, fecal microbiota transfer, farnesoid X receptor agonists, and microRNA may be potential therapeutic targets for iron overload-induced liver injury by protecting gut barrier function.
Collapse
Affiliation(s)
- Yu Liu
- Department of Health Laboratory Technology, School of Public Health, China Medical University, Shenyang, Liaoning 110122, China
| | - Guangyan Li
- Department of Health Laboratory Technology, School of Public Health, China Medical University, Shenyang, Liaoning 110122, China
| | - Fayu Lu
- School of Public Health, China Medical University, Shenyang, Liaoning 110122, China
| | - Ziwei Guo
- Department of Health Laboratory Technology, School of Public Health, China Medical University, Shenyang, Liaoning 110122, China
| | - Shuang Cai
- The First Affiliated Hospital of China Medical University, Shenyang 110001, China.
| | - Taoguang Huo
- Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China; Department of Health Laboratory Technology, School of Public Health, China Medical University, Shenyang, Liaoning 110122, China.
| |
Collapse
|
18
|
Wang C, Fu Y, Wang R, Wang Q, Yu H, Zhang J. Quercetin Attenuates the Combined Effects of Zearalenone and Lipopolysaccharide on IPEC-J2 Cell Injury through Activating the Nrf2 Signaling Pathway. Toxins (Basel) 2023; 15:679. [PMID: 38133183 PMCID: PMC10748267 DOI: 10.3390/toxins15120679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 11/09/2023] [Accepted: 11/28/2023] [Indexed: 12/23/2023] Open
Abstract
Zearalenone (ZEA) is a mycotoxin with an estrogen-like effect that is widely found in feed. Lipopolysaccharides (LPS) derived from Gram-negative bacteria are a common endotoxin, and both toxins have effects on human and livestock health. During animal feeding, ZEA as an exotoxin and LPS as an endotoxin have the potential to co-exist in organisms. At present, other studies have only focused on the hazards of single toxins, but there are fewer studies on the coexistence and interaction between ZEA and LPS. Therefore, a further study to investigate the combined toxic effects of different concentrations of ZEA and LPS is warranted. Quercetin (QUE) is a natural flavonoid compound with strong antioxidant and anti-inflammatory properties. It is unclear whether QUE can mitigate the combined effects of ZEA and LPS. IPEC-J2, isolated from the jejunum of non-breastfed neonatal piglets, is an ideal model for the study of epithelial cell transport, intestinal bacterial interactions, and the nutrient modulation of intestinal function. Therefore, the purpose of the present study was to demonstrate the effect of QUE in alleviating the combined toxic effect of ZEA and LPS on IPEC-J2 cell damage. Cell viability was measured after treating IPEC-J2 cells sequentially with 10, 20, 30, 40, 60, 80, and 100 μM ZEA, 1, 10, 50, and 100 μg/mL LPS, and 20, 40, 60, 80, 100, and 200 μM QUE for 24 h. Based on the cell viability results, 20 μM ZEA and 1 μg/mL LPS were selected as the most suitable concentrations for further analysis. For QUE, 20 μM increased the cell viability, while 40-200 μM QUE decreased the cell viability. Therefore, for the subsequent study, 20 μM QUE was selected in combination with 20 μM ZEA and 1 μg/mL LPS. The results showed that QUE increased the cellular viability and decreased the LDH content more compared to the effects of the ZEA+LPS group. At the gene level, QUE addition up-regulated the expression of Nrf2, HO-1, SOD2, and NQO1 at the gene or protein level compared to those of the ZEA+LPS group. The measurement of tight junction-related genes and proteins showed QUE up-regulated the expression of Claudin, ZO-1, and Occludin genes and proteins more than in the ZEA+LPS group. QUE addition reduced the rate of apoptosis more than that in the ZEA+LPS group. The expressions of Bcl-2 and Bax were examined at the gene level, and QUE addition significantly reduced the Bax gene expression level compared to that of the ZEA+LPS group, but there was no apparent variation in the expression level of Bcl-2. In summary, QUE can alleviate the combined toxic effects of ZEA and LPS on IPEC-J2 cells via modulating the Nrf2 signaling pathway, up-regulating the expression of antioxidative genes, and enhancing the intestinal barrier.
Collapse
Affiliation(s)
- Chuanqi Wang
- Jilin Provincial Key Laboratory of Livestock and Poultry Feed and Feeding in the Northeastern Frigid Area, College of Animal Sciences, Jilin University, Changchun 130062, China; (C.W.); (R.W.); (Q.W.); (H.Y.)
| | - Yurong Fu
- Institute of Cereal and Oil Crops, Hebei Academy of Agriculture and Forestry Sciences, Hebei Key Laboratory of Crop Cultivation Physiology and Green Production, Shijiazhuang 050035, China;
| | - Ruqi Wang
- Jilin Provincial Key Laboratory of Livestock and Poultry Feed and Feeding in the Northeastern Frigid Area, College of Animal Sciences, Jilin University, Changchun 130062, China; (C.W.); (R.W.); (Q.W.); (H.Y.)
| | - Qiyuan Wang
- Jilin Provincial Key Laboratory of Livestock and Poultry Feed and Feeding in the Northeastern Frigid Area, College of Animal Sciences, Jilin University, Changchun 130062, China; (C.W.); (R.W.); (Q.W.); (H.Y.)
| | - Hao Yu
- Jilin Provincial Key Laboratory of Livestock and Poultry Feed and Feeding in the Northeastern Frigid Area, College of Animal Sciences, Jilin University, Changchun 130062, China; (C.W.); (R.W.); (Q.W.); (H.Y.)
| | - Jing Zhang
- Jilin Provincial Key Laboratory of Livestock and Poultry Feed and Feeding in the Northeastern Frigid Area, College of Animal Sciences, Jilin University, Changchun 130062, China; (C.W.); (R.W.); (Q.W.); (H.Y.)
| |
Collapse
|
19
|
Zong Y, Meng J, Mao T, Han Q, Zhang P, Shi L. Repairing the intestinal mucosal barrier of traditional Chinese medicine for ulcerative colitis: a review. Front Pharmacol 2023; 14:1273407. [PMID: 37942490 PMCID: PMC10628444 DOI: 10.3389/fphar.2023.1273407] [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: 08/06/2023] [Accepted: 10/10/2023] [Indexed: 11/10/2023] Open
Abstract
Damage to the intestinal mucosal barrier play an important role in the pathogenesis of ulcerative colitis (UC). Discovering the key regulators and repairing the disturbed barrier are crucial for preventing and treating UC. Traditional Chinese medicine (TCM) has been proved to be effective on treating UC and has exhibited its role in repairing the intestinal mucosal barrier. We summarized the evidence of TCM against UC by protecting and repairing the physical barrier, chemical barrier, immune barrier, and biological barrier. Mechanisms of increasing intestinal epithelial cells, tight junction proteins, and mucins, promoting intestinal stem cell proliferation, restoring the abundance of the intestinal microbiota, and modulating the innate and adaptive immunity in gut, were all involved in. Some upstream proteins and signaling pathways have been elucidated. Based on the existing problems, we suggested future studies paying attention to patients' samples and animal models of UC and TCM syndromes, conducting rescue experiments, exploring more upstream regulators, and adopting new technical methods. We hope this review can provide a theoretical basis and novel ideas for clarifying the mechanisms of TCM against UC via repairing the intestinal mucosal barrier.
Collapse
Affiliation(s)
- Yichen Zong
- Second Clinical Medical College, Beijing University of Chinese Medicine, Beijing, China
| | - Jie Meng
- Department of Gastroenterology and Hepatology, Beijing University of Chinese Medicine Affiliated Dongfang Hospital, Beijing, China
| | - Tangyou Mao
- Department of Gastroenterology and Hepatology, Beijing University of Chinese Medicine Affiliated Dongfang Hospital, Beijing, China
| | - Qiang Han
- Department of Traditional Chinese Medicine, Health Service Center of Beiyuan Community, Beijing, China
| | - Peng Zhang
- Department of Gastroenterology and Hepatology, Beijing University of Chinese Medicine Affiliated Dongfang Hospital, Beijing, China
| | - Lei Shi
- Department of Gastroenterology and Hepatology, Beijing University of Chinese Medicine Affiliated Dongfang Hospital, Beijing, China
| |
Collapse
|
20
|
Tian S, Chu Q, Ma S, Ma H, Song H. Dietary Fiber and Its Potential Role in Obesity: A Focus on Modulating the Gut Microbiota. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:14853-14869. [PMID: 37815013 DOI: 10.1021/acs.jafc.3c03923] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/11/2023]
Abstract
Dietary fiber is a carbohydrate polymer with ten or more monomeric units that are resistant to digestion by human digestive enzymes, and it has gained widespread attention due to its significant role in health improvement through regulating gut microbiota. In this review, we summarized the interaction between dietary fiber, gut microbiota, and obesity, and the beneficial effects of dietary fiber on obesity through the modulation of microbiota, such as modifying selective microbial composition, producing starch-degrading enzymes, improving gut barrier function, reducing the inflammatory response, reducing trimethylamine N-oxide, and promoting the production of gut microbial metabolites (e.g., short chain fatty acids, bile acids, ferulic acid, and succinate). In addition, factors affecting the gut microbiota composition and metabolites by dietary fiber (length of the chain, monosaccharide composition, glycosidic bonds) were also concluded. Moreover, strategies for enhancing the biological activity of dietary fiber (fermentation technology, ultrasonic modification, nanotechnology, and microfluidization) were subsequently discussed. This review may provide clues for deeply exploring the structure-activity relationship between dietary fiber and antiobesity properties by targeting specific gut microbiota.
Collapse
Affiliation(s)
- Shuhua Tian
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing 210023, China
| | - Qiang Chu
- Tea Research Institute, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou 310058, PR China
| | - Shaotong Ma
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing 210023, China
| | - Huan Ma
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing 210023, China
| | - Haizhao Song
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing 210023, China
| |
Collapse
|
21
|
Mahapatro A, Bawna F, Kumar V, Daryagasht AA, Gupta S, Raghuma N, Moghdam SS, Kolla A, Mahapatra SS, Sattari N, Amini-Salehi E, Nayak SS. Anti-inflammatory effects of probiotics and synbiotics on patients with non-alcoholic fatty liver disease: An umbrella study on meta-analyses. Clin Nutr ESPEN 2023; 57:475-486. [PMID: 37739694 DOI: 10.1016/j.clnesp.2023.07.087] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 07/28/2023] [Indexed: 09/24/2023]
Abstract
BACKGROUND AND AIM The impact of chronic low-grade inflammation in the development of non-alcoholic fatty liver disease (NAFLD) has been studied widely. Previous studies showed gut pathogens' effects on inflammation development in NAFLD patients; hence, hypothetically, gut microbial therapy by administration of probiotics, synbiotics, and prebiotics may alleviate inflammation in these individuals. Several studies were performed in this regard; however, conflicting results were obtained. In this study, we aimed to comprehensively evaluate the effects of gut microbial therapy on inflammatory markers in NAFLD patients in a meta-umbrella design. METHODS Two independent researchers investigated international databases, including PubMed, Web of Science, Scopus, and Cochrane Library, from inception until March 2023. Meta-analyses evaluating the impact of probiotics, synbiotics, or prebiotics on inflammatory markers of patients with NAFLD were eligible for our study. AMASTAR 2 checklist was used to evaluate the quality of included studies. Random effect model was performed for the analysis, and Egger's regression test was conducted to determine publication bias. RESULTS A total number of 12 studies were entered into our analysis. Our findings revealed that gut microbial therapy could significantly reduce serum C-reactive protein (CRP) levels among NAFLD patients (ES: -0.58; 95% CI: -0.73, -0.44, P < 0.001). In subgroup analysis, this reduction was observed with both probiotics (ES: -0.63; 95% CI: -0.81, -0.45, P < 0.001) and synbiotics (ES: -0.49; 95% CI: -0.74, -0.24, P < 0.001). In addition, gut microbial therapy could significantly decrease tumor necrosis factor-a (TNF-a) levels in NAFLD patients (ES: -0.48; 95% CI: -0.67 to -0.30, P < 0.001). In subgroup analysis, this decrease was observed with probiotics (ES: -0.32; 95% CI: -0.53, -0.11, P = 0.002) and synbiotics (ES: -0.96; 95% CI: -1.32, -0.60, P < 0.001). Not enough information was available for assessing prebiotics' impacts. CONCLUSION The results of this umbrella review suggest that probiotics and synbiotics have promising effects on inflammatory markers, including TNF-a and CRP; however, more research is needed regarding the effects of prebiotics. PROSPERO REGISTRATION CODE CRD42022346998.
Collapse
Affiliation(s)
| | - Fnu Bawna
- Dow University of Health Sciences, Karachi, Pakistan
| | | | | | - Siddharth Gupta
- Baptist Memorial Hospital, North Mississippi, Mississippi, USA
| | - Nakka Raghuma
- GSL Medical College and General Hospital, Rajamahendravaram, Andhra Pradesh, India
| | | | - Akshita Kolla
- SRM Medical College Hospital and Research Center, Chennai, India
| | | | - Nazila Sattari
- School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | | | - Sandeep S Nayak
- Department of Internal Medicine, Bridgeport Hospital, Bridgeport, USA
| |
Collapse
|
22
|
Dai ZQ, Shang LJ, Wei YS, Li ZQ, Zeng XF, Chen MX, Wang XY, Li SY, Qiao S, Yu H. Immunomodulatory Effects of Microcin C7 in Cyclophosphamide-Induced Immunosuppressed Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:12700-12714. [PMID: 37602796 DOI: 10.1021/acs.jafc.3c01033] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/22/2023]
Abstract
Microcin C7 (McC) as a viable immunomodulator peptide can be a potential solution for pathogenic microbial infection in the post-antibiotic era and has gained substantial attention. This study was designed to evaluate the immunomodulatory activity of Microcin C7 in a cyclophosphamide (CTX)-induced immunodeficient mouse model. We show that Microcin C7 treatment significantly alleviated the CTX-caused body weight loss, improved the feed and water consumption to improve the state of the mice, and elevated the absolute number and proportion of peripheral blood lymphocytes as well as the level of hemoglobulin. We further aim to characterize the phenotypes of the immune function and intestinal health profiles. The results demonstrate that Microcin C7 treatment increased serum levels of immunoglobulin A (IgA), IgG, interleukin 6, and hemolysin, promoted splenic lymphocyte proliferation induced by concanavalin A and LPS, and enhanced the phagocytosis of peritoneal macrophages immunized by sheep red blood cells. Additionally, Microcin C7 treatment decreased levels of diamine oxidase and d-lactate, ameliorated CTX-induced intestinal morphological damage, and increased the levels of zonula occluden 1, occludin, claudin-1, mucin 2, and secretary IgA in the jejunum and colon. Moreover, Microcin C7 administration is sufficient to reverse CTX-induced intestinal microbiota dysbiosis by increasing the number of Lactobacillus and Bifidobacterium, decreasing the number of Escherichia coli in colonic contents. Collectively, our results demonstrate that Microcin C7 may have protective and immunomodulatory functions and could be a potential candidate used in animal feed, functional foods, and immunological regimens..
Collapse
Affiliation(s)
- Zi-Qi Dai
- State Key Laboratory of Animal Nutrition, Ministry of Agriculture and Rural Affairs Feed Industry Center, College of Animal Science and Technology, China Agricultural University, Beijing 100193, P. R. China
- Beijing Bio-Feed Additives Key Laboratory, Beijing 100193, China
| | - Li-Jun Shang
- State Key Laboratory of Animal Nutrition, Ministry of Agriculture and Rural Affairs Feed Industry Center, College of Animal Science and Technology, China Agricultural University, Beijing 100193, P. R. China
- Beijing Bio-Feed Additives Key Laboratory, Beijing 100193, China
| | - Yu-Shu Wei
- College of Animal Science and Technology, Shandong Agricultural University, Taian, Shandong 271018, P. R. China
| | - Ze-Qiang Li
- Luzhou Modern Agriculture Development Promotion Center, Luzhou, Sichuan 646000, P. R. China
| | - Xiang-Fang Zeng
- State Key Laboratory of Animal Nutrition, Ministry of Agriculture and Rural Affairs Feed Industry Center, College of Animal Science and Technology, China Agricultural University, Beijing 100193, P. R. China
- Beijing Bio-Feed Additives Key Laboratory, Beijing 100193, China
| | - Mei-Xia Chen
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing 100097, China
| | - Xin-Yu Wang
- State Key Laboratory of Animal Nutrition, Ministry of Agriculture and Rural Affairs Feed Industry Center, College of Animal Science and Technology, China Agricultural University, Beijing 100193, P. R. China
- Beijing Bio-Feed Additives Key Laboratory, Beijing 100193, China
| | - Si-Yu Li
- State Key Laboratory of Animal Nutrition, Ministry of Agriculture and Rural Affairs Feed Industry Center, College of Animal Science and Technology, China Agricultural University, Beijing 100193, P. R. China
- Beijing Bio-Feed Additives Key Laboratory, Beijing 100193, China
| | - Shiyan Qiao
- State Key Laboratory of Animal Nutrition, Ministry of Agriculture and Rural Affairs Feed Industry Center, College of Animal Science and Technology, China Agricultural University, Beijing 100193, P. R. China
- Beijing Bio-Feed Additives Key Laboratory, Beijing 100193, China
| | - Haitao Yu
- State Key Laboratory of Animal Nutrition, Ministry of Agriculture and Rural Affairs Feed Industry Center, College of Animal Science and Technology, China Agricultural University, Beijing 100193, P. R. China
- Beijing Bio-Feed Additives Key Laboratory, Beijing 100193, China
| |
Collapse
|
23
|
Avelar-Rodríguez D, Peña-Vélez R, Popov J, Hill L, Ryan PM. Probiotics and non-alcoholic fatty liver disease in children and adolescents: a systematic review. REVISTA ESPANOLA DE ENFERMEDADES DIGESTIVAS 2023; 115:418-427. [PMID: 36412490 DOI: 10.17235/reed.2022.8796/2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/05/2023]
Abstract
BACKGROUND non-alcoholic fatty liver disease (NAFLD) in childhood is an increasing global public health issue with significant long-term consequences. NAFLD management mainly consists of lifestyle modifications, however, adjunct pharmacological therapies are currently lacking. Gut microbiota manipulation via probiotics may alter the course of pediatric NAFLD. The objective of this systematic review was to synthesize all the available literature on the use of probiotics in children and adolescents with NAFLD. METHODS PubMed, EBSCOhost, Scopus, Web of Science, and Cochrane Library were systematically searched for trials on the use of probiotics in pediatric NAFLD. A quantitative DerSimonian Laird random effects meta-analysis was performed when possible; otherwise, a narrative summary of the study outcomes was presented and discussed. A separate search was completed to include all the ongoing registered trials on probiotics use in pediatric NAFLD. RESULTS five randomized controlled trials met the inclusion criteria. Of these, four trials were included in the final quantitative analysis. Probiotic therapy significantly reduced the levels of alanine aminotransferase (ALT) (mean difference: -10.39 [-19.85, -0.93]), however significant heterogeneity between studies was identified (I2, 93 %). CONCLUSIONS there is insufficient evidence to support probiotics in the treatment of pediatric NAFLD given the substantial degree of discordance amongst the available trials. Lifestyle modifications focusing on maintaining a normal BMI and regular exercise continue to be the gold standard approach to treating NAFLD in children.
Collapse
Affiliation(s)
| | - Rubén Peña-Vélez
- Gastroenterología y Nutrición, Hospital General de Puebla "Dr. Eduardo Vázquez N", México
| | - Jelena Popov
- College of Medicine and Health. University College Cork
| | | | | |
Collapse
|
24
|
Zeebone YY, Bóta B, Halas V, Libisch B, Olasz F, Papp P, Keresztény T, Gerőcs A, Ali O, Kovács M, Szabó A. Gut-Faecal Microbial and Health-Marker Response to Dietary Fumonisins in Weaned Pigs. Toxins (Basel) 2023; 15:328. [PMID: 37235363 PMCID: PMC10222793 DOI: 10.3390/toxins15050328] [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: 02/15/2023] [Revised: 04/07/2023] [Accepted: 05/06/2023] [Indexed: 05/28/2023] Open
Abstract
This study investigated effects of dietary fumonisins (FBs) on gut and faecal microbiota of weaned pigs. In total, 18 7-week-old male pigs were fed either 0, 15 or 30 mg FBs (FB1 + FB2 + FB3)/kg diet for 21 days. The microbiota was analysed with amplicon sequencing of the 16S rRNA gene V3-V4 regions (Illumina MiSeq). Results showed no treatment effect (p > 0.05) on growth performance, serum reduced glutathione, glutathione peroxidase and malondialdehyde. FBs increased serum aspartate transaminase, gamma glutamyl-transferase and alkaline phosphatase activities. A 30 mg/kg FBs treatment shifted microbial population in the duodenum and ileum to lower levels (compared to control (p < 0.05)) of the families Campylobacteraceae and Clostridiaceae, respectively, as well as the genera Alloprevotella, Campylobacter and Lachnospiraceae Incertae Sedis (duodenum), Turicibacter (jejunum), and Clostridium sensu stricto 1 (ileum). Faecal microbiota had higher levels of the Erysipelotrichaceae and Ruminococcaceae families and Solobacterium, Faecalibacterium, Anaerofilum, Ruminococcus, Subdoligranulum, Pseudobutyrivibrio, Coprococcus and Roseburia genera in the 30 mg/kg FBs compared to control and/or to the 15 mg/kg FBs diets. Lactobacillus was more abundant in the duodenum compared to faeces in all treatment groups (p < 0.01). Overall, the 30 mg/kg FBs diet altered the pig gut microbiota without suppressing animal growth performance.
Collapse
Affiliation(s)
- Yarsmin Yunus Zeebone
- Agribiotechnology and Precision Breeding for Food Security National Laboratory, Institute of Physiology and Nutrition, Hungarian University of Agriculture and Life Sciences, Guba S. Str., H-7400 Kaposvár, Hungary
- ELKH-MATE Mycotoxins in the Food Chain Research Group, Hungarian University of Agriculture and Life Sciences, Guba S. Str., H-7400 Kaposvár, Hungary
| | - Brigitta Bóta
- ELKH-MATE Mycotoxins in the Food Chain Research Group, Hungarian University of Agriculture and Life Sciences, Guba S. Str., H-7400 Kaposvár, Hungary
| | - Veronika Halas
- Department of Farm Animal Nutrition, Institute of Animal Physiology and Nutrition, Kaposvár Campus, Hungarian University of Agriculture and Life Sciences, Guba S. Str., H-7400 Kaposvár, Hungary
| | - Balázs Libisch
- Agribiotechnology and Precision Breeding for Food Security National Laboratory, Institute of Genetics and Biotechnology, Hungarian University of Agriculture and Life Sciences, Szent-Györgyi. Str., H-2100 Gödöllő, Hungary
| | - Ferenc Olasz
- Agribiotechnology and Precision Breeding for Food Security National Laboratory, Institute of Genetics and Biotechnology, Hungarian University of Agriculture and Life Sciences, Szent-Györgyi. Str., H-2100 Gödöllő, Hungary
| | - Péter Papp
- Agribiotechnology and Precision Breeding for Food Security National Laboratory, Institute of Genetics and Biotechnology, Hungarian University of Agriculture and Life Sciences, Szent-Györgyi. Str., H-2100 Gödöllő, Hungary
| | - Tibor Keresztény
- Agribiotechnology and Precision Breeding for Food Security National Laboratory, Institute of Genetics and Biotechnology, Hungarian University of Agriculture and Life Sciences, Szent-Györgyi. Str., H-2100 Gödöllő, Hungary
- Doctoral School of Biology, Hungarian University of Agriculture and Life Sciences, H-2100 Gödöllő, Hungary
| | - Annamária Gerőcs
- Agribiotechnology and Precision Breeding for Food Security National Laboratory, Institute of Genetics and Biotechnology, Hungarian University of Agriculture and Life Sciences, Szent-Györgyi. Str., H-2100 Gödöllő, Hungary
- Doctoral School of Biology, ELTE Eötvös Loránd University, H-1117 Budapest, Hungary
| | - Omeralfaroug Ali
- Agribiotechnology and Precision Breeding for Food Security National Laboratory, Institute of Physiology and Nutrition, Hungarian University of Agriculture and Life Sciences, Guba S. Str., H-7400 Kaposvár, Hungary
- ELKH-MATE Mycotoxins in the Food Chain Research Group, Hungarian University of Agriculture and Life Sciences, Guba S. Str., H-7400 Kaposvár, Hungary
| | - Melinda Kovács
- Agribiotechnology and Precision Breeding for Food Security National Laboratory, Institute of Physiology and Nutrition, Hungarian University of Agriculture and Life Sciences, Guba S. Str., H-7400 Kaposvár, Hungary
- ELKH-MATE Mycotoxins in the Food Chain Research Group, Hungarian University of Agriculture and Life Sciences, Guba S. Str., H-7400 Kaposvár, Hungary
| | - András Szabó
- Agribiotechnology and Precision Breeding for Food Security National Laboratory, Institute of Physiology and Nutrition, Hungarian University of Agriculture and Life Sciences, Guba S. Str., H-7400 Kaposvár, Hungary
- ELKH-MATE Mycotoxins in the Food Chain Research Group, Hungarian University of Agriculture and Life Sciences, Guba S. Str., H-7400 Kaposvár, Hungary
| |
Collapse
|
25
|
Wang Y, Ren K, Tan J, Mao Y. Alginate oligosaccharide alleviates aging-related intestinal mucosal barrier dysfunction by blocking FGF1-mediated TLR4/NF-κB p65 pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 116:154806. [PMID: 37236046 DOI: 10.1016/j.phymed.2023.154806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 03/30/2023] [Accepted: 04/04/2023] [Indexed: 05/28/2023]
Abstract
BACKGROUND Alginate oligosaccharide (AOS) has been reported to exert a crucial role in maintaining the intestinal mucosal barrier (IMB) function. The current study aimed at ascertaining the protective effects of AOS on aging-induced IMB dysfunction and to elucidate the underlying molecular mechanisms. METHODS An aging mouse model and a senescent NCM460 cell model were established using d-galactose. AOS was administered to aging mice and senescent cells, and IMB permeability, inflammatory response and tight junction proteins were assessed. In silico analysis was conducted to identify factors regulated by AOS. Using gain- and loss-of-function approaches, we evaluated the roles of FGF1, TLR4 and NF-κB p65 in the aging-induced IMB dysfunction and NCM460 cell senescence. RESULTS AOS protected the IMB function of aging mice and NCM460 cells by reducing permeability and increasing tight junction proteins. In addition, AOS up-regulated FGF1, which blocked the TLR4/NF-κB p65 pathway, and identified as the mechanism responsible for the protective effect of AOS. CONCLUSION AOS blocks the TLR4/NF-κB p65 pathway via inducing FGF1, ultimately reducing the risk of IMB dysfunction in aging mice. This study highlights the potential of AOS as a protective agent against aging-induced IMB disorder and provides insight into the underlying molecular mechanisms.
Collapse
Affiliation(s)
- Yanting Wang
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao, Shandong 266000, China
| | - Keyu Ren
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao, Shandong 266000, China
| | - Junying Tan
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao, Shandong 266000, China
| | - Yongjun Mao
- Department of Geriatric Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266000, China.
| |
Collapse
|
26
|
Zhou F, Lin Y, Chen S, Bao X, Fu S, Lv Y, Zhou M, Chen Y, Zhu B, Qian C, Li Z, Ding Z. Ameliorating role of Tetrastigma hemsleyanum polysaccharides in antibiotic-induced intestinal mucosal barrier dysfunction in mice based on microbiome and metabolome analyses. Int J Biol Macromol 2023; 241:124419. [PMID: 37080409 DOI: 10.1016/j.ijbiomac.2023.124419] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 04/03/2023] [Accepted: 04/08/2023] [Indexed: 04/22/2023]
Abstract
The intestinal mucosal barrier is one of the important barriers to prevent harmful substances and pathogens from entering the body environment and to maintain intestinal homeostasis. This study investigated the reparative effect and possible mechanism of Tetrastigma hemsleyanum polysaccharides (THP) on ceftriaxone-induced intestinal mucosal damage. Our results suggested that THP repaired the mechanical barrier damage of intestinal mucosa by enhancing the expression of intestinal tight junction proteins, reducing intestinal mucosal permeability and improving the pathological state of intestinal epithelial cells. Intestinal immune and chemical barrier was further restored by THP via the increment of the body's cytokine levels, intestinal SIgA levels, intestinal goblet cell number, intestinal mucin-2 levels, and short-chain fatty acid levels. In addition, THP increased the abundance of probiotic bacteria (such as Lactobacillus), reduced the abundance of harmful bacteria (such as Enterococcus) to repair the intestinal biological barrier, restored intestinal mucosal barrier function, and maintains intestinal homeostasis. The possible mechanisms were related to sphingolipid metabolism, linoleic acid metabolism, and d-glutamine and D-glutamate metabolism. Our results demonstrated the potential therapeutic effect of THP against intestinal flora disorders and intestinal barrier function impairment caused by antibiotics.
Collapse
Affiliation(s)
- Fangmei Zhou
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China
| | - Yue Lin
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China
| | - Senmiao Chen
- College of Life Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China
| | - Xiaodan Bao
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China
| | - Siyu Fu
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China
| | - Yishan Lv
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China
| | - Mingyuan Zhou
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China
| | - Yuchi Chen
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China
| | - Bingqi Zhu
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China
| | - Chaodong Qian
- College of Life Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China
| | - Zhimin Li
- Information Technology Center, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China
| | - Zhishan Ding
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China.
| |
Collapse
|
27
|
Pan Z, Huang J, Hu T, Zhang Y, Zhang L, Zhang J, Cui D, Li L, Wang J, Wu Q. Protective Effects of Selenium Nanoparticles against Bisphenol A-Induced Toxicity in Porcine Intestinal Epithelial Cells. Int J Mol Sci 2023; 24:ijms24087242. [PMID: 37108405 PMCID: PMC10139072 DOI: 10.3390/ijms24087242] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 04/05/2023] [Accepted: 04/09/2023] [Indexed: 04/29/2023] Open
Abstract
Bisphenol A (BPA) is widely used to harden plastics and polycarbonates and causes serious toxic effects in multiple organs, including the intestines. Selenium, as an essential nutrient element for humans and animals, exhibits a predominant effect in various physiological processes. Selenium nanoparticles have attracted more and more attention due to their outstanding biological activity and biosafety. We prepared chitosan-coated selenium nanoparticles (SeNPs) and further compared the protective effects, and investigated the underlying mechanism of SeNPs and inorganic selenium (Na2SeO3) on BPA-induced toxicity in porcine intestinal epithelial cells (IPEC-J2). The particle size, zeta potential, and microstructure of SeNPs were detected by using a nano-selenium particle size meter and a transmission electron microscope. IPEC-J2 cells were exposed to BPA alone or simultaneously exposed to BPA and SeNPs or Na2SeO3. The CCK8 assay was performed to screen the optimal concentration of BPA exposure and the optimal concentration of SeNPs and Na2SeO3 treatment. The apoptosis rate was detected by flow cytometry. Real-time PCR and Western blot methods were used to analyze the mRNA and protein expression of factors related to tight junctions, apoptosis, inflammatory responses and endoplasmic reticulum stress. Increased death and morphological damage were observed after BPA exposure, and these increases were attenuated by SeNPs and Na2SeO3 treatment. BPA exposure disturbed the tight junction function involved with decreased expression of tight junction protein Zonula occludens 1 (ZO-1), occludin, and claudin-1 proteins. Proinflammatory response mediated by the transcription factor nuclear factor-k-gene binding (NF-κB), such as elevated levels of interleukin-1β(IL-1β), interleukin-6 (IL-6), interferon-γ (IFN-γ), interleukin-17 (IL-17), and tumor necrosis factor-α (TNF-α) expression was induced at 6 and 24 h after BPA exposure. BPA exposure also disturbed the oxidant/antioxidant status and led to oxidative stress. IPEC-J2 cell apoptosis was induced by BPA exposure, as indicated by increased BCL-2-associated X protein (Bax), caspase 3, caspase 8, and caspase 9 expression and decreased B-cell lymphoma-2 (Bcl-2) and Bcl-xl expression. BPA exposure activated the endoplasmic reticulum stress (ERS) mediated by the receptor protein kinase receptor-like endoplasmic reticulum kinase (PERK), Inositol requiring enzyme 1 (IRE1α), and activating transcription factor 6 (ATF6). We found that treatment with SeNPs and Na2SeO3 can alleviate the intestinal damage caused by BPA. SeNPs were superior to Na2SeO3 and counteracted BPA-induced tight junction function injury, proinflammatory response, oxidative stress, apoptosis, and ERS stress. Our findings suggest that SeNPs protect intestinal epithelial cells from BPA-induced damage, partly through inhibiting ER stress activation and subsequently attenuating proinflammatory responses and oxidative stress and suppressing apoptosis, thus enhancing the intestinal epithelial barrier function. Our data indicate that selenium nanoparticles may represent an effective and reliable tool for preventing BPA toxicity in animals and humans.
Collapse
Affiliation(s)
- Zaozao Pan
- Animal Science and Technology College, Beijing University of Agriculture, Beijing 100096, China
| | - Jiaqiang Huang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China
| | - Ting Hu
- Animal Science and Technology College, Beijing University of Agriculture, Beijing 100096, China
| | - Yonghong Zhang
- Animal Science and Technology College, Beijing University of Agriculture, Beijing 100096, China
| | - Lingyu Zhang
- Animal Science and Technology College, Beijing University of Agriculture, Beijing 100096, China
| | - Jiaxi Zhang
- Animal Science and Technology College, Beijing University of Agriculture, Beijing 100096, China
| | - Defeng Cui
- Animal Science and Technology College, Beijing University of Agriculture, Beijing 100096, China
| | - Lu Li
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing 100097, China
| | - Jing Wang
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing 100097, China
| | - Qiong Wu
- Animal Science and Technology College, Beijing University of Agriculture, Beijing 100096, China
| |
Collapse
|
28
|
Xiang X, Wang X, Shang Y, Ding Y. Microfluidic intestine-on-a-chip: Current progress and further perspectives of probiotic-foodborne pathogen interactions. Trends Food Sci Technol 2023. [DOI: 10.1016/j.tifs.2023.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023]
|
29
|
Ji J, Wu L, Wei J, Wu J, Guo C. The Gut Microbiome and Ferroptosis in MAFLD. J Clin Transl Hepatol 2023; 11:174-187. [PMID: 36406312 PMCID: PMC9647110 DOI: 10.14218/jcth.2022.00136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 05/22/2022] [Accepted: 06/12/2022] [Indexed: 12/04/2022] Open
Abstract
Metabolic-associated fatty liver disease (MAFLD) is a new disease definition, and is proposed to replace the previous name, nonalcoholic fatty liver disease (NAFLD). Globally, MAFLD/NAFLD is the most common liver disease, with an incidence rate ranging from 6% to 35% in adult populations. The pathogenesis of MAFLD/NAFLD is closely related to insulin resistance (IR), and the genetic susceptibility to acquired metabolic stress-associated liver injury. Similarly, the gut microbiota in MAFLD/NAFLD is being revaluated by scientists, as the gut and liver influence each other via the gut-liver axis. Ferroptosis is a novel form of programmed cell death caused by iron-dependent lipid peroxidation. Emerging evidence suggests that ferroptosis has a key role in the pathological progression of MAFLD/NAFLD, and inhibition of ferroptosis may become a novel therapeutic strategy for the treatment of NAFLD. This review focuses on the main mechanisms behind the promotion of MAFLD/NAFLD occurrence and development by the intestinal microbiota and ferroptosis. It outlines new strategies to target the intestinal microbiota and ferroptosis to facilitate future MAFLD/NAFLD therapies.
Collapse
Affiliation(s)
- Jie Ji
- Department of Gastroenterology, Putuo People’s Hospital, Tongji University, Shanghai, China
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Liwei Wu
- Department of Gastroenterology, Putuo People’s Hospital, Tongji University, Shanghai, China
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jue Wei
- Department of Gastroenterology Shanghai Tongren Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jianye Wu
- Department of Gastroenterology, Putuo People’s Hospital, Tongji University, Shanghai, China
- Correspondence to: Chuanyong Guo, Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, NO. 301, Middle Yanchang Road, Jing’an District, Shanghai 200072, China. ORCID: https://orcid.org/0000-0002-6527-4673. E-mail: ; Jianye Wu: Department of Gastroenterology, Putuo People’s Hospital, NO. 1291, Jiangning road, Putuo, Shanghai 200060, China. ORCID: https://orcid.org/0000-0003-2675-4241. E-mail:
| | - Chuanyong Guo
- Department of Gastroenterology, Putuo People’s Hospital, Tongji University, Shanghai, China
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
- Correspondence to: Chuanyong Guo, Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, NO. 301, Middle Yanchang Road, Jing’an District, Shanghai 200072, China. ORCID: https://orcid.org/0000-0002-6527-4673. E-mail: ; Jianye Wu: Department of Gastroenterology, Putuo People’s Hospital, NO. 1291, Jiangning road, Putuo, Shanghai 200060, China. ORCID: https://orcid.org/0000-0003-2675-4241. E-mail:
| |
Collapse
|
30
|
Liu S, Li J, Kang W, Li Y, Ge L, Liu D, Liu Y, Huang K. Aflatoxin B1 Induces Intestinal Barrier Dysfunction by Regulating the FXR-Mediated MLCK Signaling Pathway in Mice and in IPEC-J2 Cells. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:867-876. [PMID: 36579420 DOI: 10.1021/acs.jafc.2c06931] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Aflatoxin B1 (AFB1) is a widespread mycotoxin in food and feed. Although the liver is the main target organ of AFB1, the intestine is the first exposure organ to AFB1. However, the mechanism by which AFB1 induced intestinal barrier dysfunction via regulating the farnesoid X receptor (FXR)-mediated myosin light chain kinase (MLCK) signaling pathway has rarely been studied. In vivo, AFB1 exposure significantly decreased the small intestine length and increased the intestinal permeability. Meanwhile, AFB1 exposure markedly suppressed the protein expressions of FXR, ZO-1, occludin, and claudin-1 and enhanced the protein expression of MLCK. In vitro, AFB1 exposure induced intestinal barrier dysfunction by the elevation in the FITC-Dextran 4 kDa flux and inhibition in the transepithelial electrical resistance in a dose-dependent manner. In addition, AFB1 exposure downregulated the mRNA and protein expressions of FXR, ZO-1, occludin, and claudin-1, redistributed the ZO-1 protein, and enhanced the protein expressions of MLCK and p-MLC. However, fexaramine (Fex, FXR agonist) pretreatment markedly reversed the AFB1-induced FXR activity reduction, MLCK protein activation, and intestinal barrier impairment in vitro and in vivo. Moreover, pretreatment with the inhibition of MLCK with ML-7 significantly alleviated the AFB1-induced intestinal barrier dysfunction and tight junction disruption in vitro. In conclusion, AFB1 induced intestinal barrier impairment via regulating the FXR-mediated MLCK signaling pathway in vitro and in vivo and provided novel insights to prevent mycotoxin poisoning in the intestine.
Collapse
Affiliation(s)
- Shuiping Liu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China
- Institute of Nutritional and Metabolic Disorders in Domestic Animals and Fowls, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China
| | - Jinyan Li
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China
- Institute of Nutritional and Metabolic Disorders in Domestic Animals and Fowls, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China
| | - Weili Kang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China
- Institute of Nutritional and Metabolic Disorders in Domestic Animals and Fowls, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China
| | - Yun Li
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China
| | - Lei Ge
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China
- Institute of Nutritional and Metabolic Disorders in Domestic Animals and Fowls, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China
| | - Dandan Liu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China
- Institute of Nutritional and Metabolic Disorders in Domestic Animals and Fowls, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China
| | - Yunhuan Liu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China
- Institute of Nutritional and Metabolic Disorders in Domestic Animals and Fowls, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China
| | - Kehe Huang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China
- Institute of Nutritional and Metabolic Disorders in Domestic Animals and Fowls, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China
| |
Collapse
|
31
|
Fan Y, Liang L, Tang X, Zhu J, Mu L, Wang M, Huang X, Gong S, Xu J, Liu T, Zhang T. Changes in the gut microbiota structure and function in rats with doxorubicin-induced heart failure. Front Cell Infect Microbiol 2023; 13:1135428. [PMID: 37180435 PMCID: PMC10173310 DOI: 10.3389/fcimb.2023.1135428] [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: 12/31/2022] [Accepted: 04/11/2023] [Indexed: 05/16/2023] Open
Abstract
Objectives The rat model of heart failure (HF) induced by doxorubicin (DOX), a broad spectrum and highly effective chemotherapeutic anthracycline with high-affinity to myocardial tissue that causes severe dose-dependent irreversible cardiotoxicity has been widely recognized and applied in HF pathogenesis and drug therapy studies. The gut microbiota (GM) has attracted significant attention due to its potential role in HF, and research in this area may provide beneficial therapeutic strategies for HF. Considering the differences in the route, mode, and total cumulative dose of DOX administration used to establish HF models, the optimal scheme for studying the correlation between GM and HF pathogenesis remains to be determined. Therefore, focusing on establishing the optimal scheme, we evaluated the correlation between GM composition/function and DOX-induced cardiotoxicity (DIC). Methods Three schemes were investigated: DOX (at total cumulative doses of 12, 15 or 18 mg/kg using a fixed or alternating dose via a tail vein or intraperitoneal injection) was administered to Sprague Dawley (SD) for six consecutive weeks. The M-mode echocardiograms performed cardiac function evaluation. Pathological changes in the intestine were observed by H&E staining and in the heart by Masson staining. The serum levels of N-terminal pre-B-type natriuretic peptide (NT-proBNP) and cardiac troponin I (cTnI) were measured by ELISA. The GM was analysed by 16S rRNA gene sequencing. Key findings Strikingly, based on the severity of cardiac dysfunction, there were marked differences in the abundance and grouping of GM under different schemes. The HF model established by tail vein injection of DOX (18 mg/kg, alternating doses) was more stable; moreover, the degree of myocardial injury and microbial composition were more consistent with the clinical manifestations of HF. Conclusions The model of HF established by tail vein injection of doxorubicin, administered at 4mg/kg body weight (2mL/kg) at weeks 1, 3 and 5, and at 2mg/kg body weight (1mL/kg) at weeks 2, 4 and 6, with a cumulative total dose of 18mg/kg, is a better protocol to study the correlation between HF and GM.
Collapse
Affiliation(s)
- Yawen Fan
- Department of Cardiovascular Diseases, The Sixth Clinical Medical School of Guangzhou University of Chinese Medicine, Shenzhen Hospital of Guangzhou University of Chinese Medicine (Futian), Shenzhen, China
| | - Lichang Liang
- Department of Preventive Treatment, The Sixth Clinical Medical School of Guangzhou University of Chinese Medicine, Shenzhen Hospital of Guangzhou University of Chinese Medicine (Futian), Shenzhen, China
| | - Xinzheng Tang
- Department of Cardiovascular Diseases, The Sixth Clinical Medical School of Guangzhou University of Chinese Medicine, Shenzhen Hospital of Guangzhou University of Chinese Medicine (Futian), Shenzhen, China
| | - Jinxian Zhu
- Department of Cardiovascular Diseases, The Sixth Clinical Medical School of Guangzhou University of Chinese Medicine, Shenzhen Hospital of Guangzhou University of Chinese Medicine (Futian), Shenzhen, China
| | - Lei Mu
- Department of Encephalopathy Diseases, Shenzhen Hospital of Beijing University of Chinese Medicine (Longgang), Shenzhen, China
| | - Mengni Wang
- Department of Cardiovascular Diseases, The Sixth Clinical Medical School of Guangzhou University of Chinese Medicine, Shenzhen Hospital of Guangzhou University of Chinese Medicine (Futian), Shenzhen, China
| | - Xuecheng Huang
- Department of Spinal Surgery, The Sixth Clinical Medical School of Guangzhou University of Chinese Medicine, Shenzhen Hospital of Guangzhou University of Chinese Medicine (Futian), Shenzhen, China
| | - Shenglan Gong
- Department of Cardiovascular Diseases, The Sixth Clinical Medical School of Guangzhou University of Chinese Medicine, Shenzhen Hospital of Guangzhou University of Chinese Medicine (Futian), Shenzhen, China
| | - Jinghan Xu
- Department of Endocrinology, The Sixth Clinical Medical School of Guangzhou University of Chinese Medicine, Shenzhen Hospital of Guangzhou University of Chinese Medicine (Futian), Shenzhen, China
| | - Tianjiao Liu
- Department of Endocrinology, The Sixth Clinical Medical School of Guangzhou University of Chinese Medicine, Shenzhen Hospital of Guangzhou University of Chinese Medicine (Futian), Shenzhen, China
| | - Tianfeng Zhang
- Department of Cardiovascular Diseases, The Sixth Clinical Medical School of Guangzhou University of Chinese Medicine, Shenzhen Hospital of Guangzhou University of Chinese Medicine (Futian), Shenzhen, China
- *Correspondence: Tianfeng Zhang,
| |
Collapse
|
32
|
Liu JP, Wang J, Zhou SX, Huang DC, Qi GH, Chen GT. Ginger polysaccharides enhance intestinal immunity by modulating gut microbiota in cyclophosphamide-induced immunosuppressed mice. Int J Biol Macromol 2022; 223:1308-1319. [PMID: 36395935 DOI: 10.1016/j.ijbiomac.2022.11.104] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 11/10/2022] [Accepted: 11/10/2022] [Indexed: 11/16/2022]
Abstract
In this study, the immunity-enhancing effect of ginger polysaccharides UGP1 and UGP2 on CTX-induced immunosuppressed mice was evaluated. The results showed that ginger polysaccharide could effectively alleviate the symptoms of weight loss and dietary intake reduction induced by CTX, increase fecal water content, reduce fecal pH, and protect immune organs of immunosuppressed mice. In addition, ginger polysaccharides also stimulated the secretion of cytokines IL-2, IL-4, TNF-α and immunoglobulin Ig-G in the serum of mice, increased the expression of Occludin and Claudin-1, and restored the level of short-chain fatty acids in the intestine to improve immune deficiency. Furthermore, ginger polysaccharides significantly reduced the relative abundance ratio of the Firmicutes and Bacteroidetes in mice and increased the relative abundance of Verrucomicrobia and Bacteroidetes at the phylum level. At the family level, ginger polysaccharides increased the relative abundance of beneficial bacteria such as Muribaculaceae, Bacteroidaceae and Lactobacillaceae, and decreased the relative abundance of harmful bacteria such as Rikenellaceae and Lachnospiraceae. Spearman correlation analysis indicated that ginger polysaccharides could enhance intestinal immunity by modulating gut microbiota associated with immune function. These results indicated that ginger polysaccharides have the potential to be a functional food ingredients or a natural medicine for the treatment of intestinal barrier injury.
Collapse
Affiliation(s)
- Jun-Ping Liu
- College of Engineering/National R&D Center for Chinese Herbal Medicine Processing, China Pharmaceutical University, Nanjing, 211198, China
| | - Jie Wang
- College of Engineering/National R&D Center for Chinese Herbal Medicine Processing, China Pharmaceutical University, Nanjing, 211198, China
| | - Si-Xuan Zhou
- College of Engineering/National R&D Center for Chinese Herbal Medicine Processing, China Pharmaceutical University, Nanjing, 211198, China
| | - De-Chun Huang
- College of Engineering/National R&D Center for Chinese Herbal Medicine Processing, China Pharmaceutical University, Nanjing, 211198, China
| | - Guo-Hong Qi
- College of Engineering/National R&D Center for Chinese Herbal Medicine Processing, China Pharmaceutical University, Nanjing, 211198, China.
| | - Gui-Tang Chen
- College of Engineering/National R&D Center for Chinese Herbal Medicine Processing, China Pharmaceutical University, Nanjing, 211198, China.
| |
Collapse
|
33
|
Song X, Qiao L, Chang J, Dou X, Zhang X, Pi S, Xu C. Dietary supplementation with selenium nanoparticles-enriched Lactobacillus casei ATCC 393 alleviates intestinal barrier dysfunction of mice exposed to deoxynivalenol by regulating endoplasmic reticulum stress and gut microbiota. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 248:114276. [PMID: 36371888 DOI: 10.1016/j.ecoenv.2022.114276] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 10/29/2022] [Accepted: 11/05/2022] [Indexed: 06/16/2023]
Abstract
Deoxynivalenol (DON), a secondary product of Fusarium metabolism, is common in wheat, corn, barley and other grain crops, posing a variety of adverse effects to environment, food safety, human and animal health. The absorption of DON mainly occurs in the proximal part of the small intestine, which can induce intestinal mucosal epithelial injury, and ultimately affect the growth performance and production performance of animals. This study was conducted to investigate the protective effects of selenium nanoparticles (SeNPs)-enriched Lactobacillus casei ATCC 393 (L. casei ATCC 393) on intestinal barrier function of C57BL/6 mice exposed to DON and its association with endoplasmic reticulum stress (ERS) and gut microbiota. The results showed that DON exposure increased the levels of interleukin-6 (IL-6) and interleukin-8 (IL-8), decreased the levels of interleukin-10 (IL-10) and transforming growth factor beta (TGF-β), caused a redox imbalance and intestinal barrier dysfunction, decreased the mRNA levels of endoplasmic reticulum- resident selenoproteins, activated ERS-protein kinase R-like endoplasmic reticulum kinase (PERK) signaling pathway, altered the composition of the gut microbiota and decreased short-chain fatty acids (SCFAs) content. Dietary supplementation with SeNPs-enriched L. casei ATCC 393 can effectively protect the integrity of intestinal barrier function by reducing inflammatory response, enhancing the antioxidant capacity, up-regulating the mRNA levels of endoplasmic reticulum-resident selenoproteins, inhibiting the activation of PERK signaling pathway, reversing gut microbiota dysbiosis and increasing the content of SCFAs in mice exposed to DON. In conclusion, dietary supplementation with SeNPs-enriched L. casei ATCC 393 effectively alleviated intestinal barrier dysfunction induced by DON in C57BL/6 mice, which may be closely associated with the regulation of ERS and gut microbiota.
Collapse
Affiliation(s)
- Xiaofan Song
- The Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China
| | - Lei Qiao
- The Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China
| | - Jiajing Chang
- The Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China
| | - Xina Dou
- The Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China
| | - Xinyi Zhang
- The Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China
| | - Shanyao Pi
- The Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China
| | - Chunlan Xu
- The Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China.
| |
Collapse
|
34
|
He Y, Li Z, Xu T, Luo D, Chi Q, Zhang Y, Li S. Polystyrene nanoplastics deteriorate LPS-modulated duodenal permeability and inflammation in mice via ROS drived-NF-κB/NLRP3 pathway. CHEMOSPHERE 2022; 307:135662. [PMID: 35830933 DOI: 10.1016/j.chemosphere.2022.135662] [Citation(s) in RCA: 64] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 07/04/2022] [Accepted: 07/07/2022] [Indexed: 06/15/2023]
Abstract
The widespread occurrence of nanoplastics (NPs), has markedly affected the ecosystem and has become a global threat to animals and human health. There is growing evidence showing that polystyrene nanoparticles (PSNPs) exposure induced enteritis and the intestinal barrier disorder. Lipopolysaccharide (LPS) can trigger the inflammation burden of various tissues. Whether PSNPs deteriorate LPS-induced intestinal damage via ROS drived-NF-κB/NLRP3 pathway is remains unknown. In this study, PSNPs exposure/PSNPs and LPS co-exposure mice model were duplicated by intraperitoneal injection. The results showed that exposure to PSNPs/LPS caused duodenal inflammation and increased permeability. We evaluated the change of duodenum structure, oxidative stress parameters, inflammatory factors, and tight junction protein in the duodenum. We found that PSNPs/LPS could aggravate the production of ROS and oxidative stress in cells, activate NF-κB/NLRP3 pathway, decrease the expression tight junction proteins (ZO-1, Claudin 1, and Occludin) levels, promote inflammatory factors (TNF-α, IL-6, and IFN-γ) expressions. Duodenal oxidative stress and inflammation in PS + LPS group were more serious than those in single exposure group, which could be alleviated by NF-kB inhibitor QNZ. Collectively, the results verified that PSNPs deteriorated LPS-induced inflammation and increasing permeability in mice duodenum via ROS drived-NF-κB/NLRP3 pathway. The current study indicated the relationship and molecular mechanism between PSNPs and intestinal injury, providing novel insights into the adverse effects of PSNPs exposure on mammals and humans.
Collapse
Affiliation(s)
- Yujiao He
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China
| | - Zhe Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China
| | - Tong Xu
- College of Chemistry, Jilin University, Changchun, 130012, PR China
| | - Dongliu Luo
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China
| | - Qianru Chi
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China
| | - Yiming Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China
| | - Shu Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China.
| |
Collapse
|
35
|
Li A, Hao W, Guan S, Wang J, An G. Mycotoxin contamination in feeds and feed materials in China in year 2020. Front Vet Sci 2022; 9:1016528. [PMID: 36299638 PMCID: PMC9589091 DOI: 10.3389/fvets.2022.1016528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
A survey of mycotoxin contamination in feed commodities in China was performed and the regional differences of mycotoxin contamination in new season corn was assessed during January 2020-November 2020 in this research. 1,610 samples were analyzed for the major mycotoxins, namely aflatoxins, zearalenone (ZEN), trichothecenes type B, fumonisins (FUM), fusariotoxin T-2 (T-2) and ochratoxin A (OTA) using methods of liquid chromatography-tandem mass spectrometry and enzyme linked immunosorbent assay. Generally, aflatoxins occurred in 16% of all samples, and ZEN, trichothecenes type B and FUM were more prevalent with positive rates of 47, 72, and 63%, respectively. T2 and OTA were rarely detected. In new season corn, samples were also seriously contaminated with ZEN, trichothecenes type B, and FUM at positive rates of 47, 76, and 79%, respectively, and their averages of positives were 112, 735, and 3,811 μg/kg, respectively. The patterns of mycotoxin occurrence showed distinct regional trends in new season corn samples. Samples from Shandong province were highly contaminated with FUM, while special attention should be paid to aflatoxins in Anhui and Jiangsu provinces of East China. The contents of trichothecenes type B and ZEN from northern to southern provinces showed downward trends. In new season corm, co-occurrence of mycotoxins was widespread, and combinations of ZEN, trichothecenes type B, and FUM were frequently observed in this study. Trichothecenes type B and ZEN concentrations showed a positive correlation coefficient of 0.294, suggesting that toxicological interactions of these toxins deserve attention.
Collapse
Affiliation(s)
- Anping Li
- Department of Animal Nutrition and Health, DSM (China) Co., Ltd., Shanghai, China
| | - Wei Hao
- Department of Animal Nutrition and Health, DSM (China) Co., Ltd., Shanghai, China
| | - Shu Guan
- Department of Animal Nutrition and Health, DSM Singapore Industrial Pte Ltd., Singapore, Singapore,*Correspondence: Shu Guan
| | - Jinyong Wang
- Department of Animal Nutrition and Health, DSM (China) Co., Ltd., Shanghai, China
| | - Gang An
- Department of Animal Nutrition and Health, DSM (China) Co., Ltd., Shanghai, China
| |
Collapse
|
36
|
Wang L, Wang X, Chang J, Wang P, Liu C, Yuan L, Yin Q, Zhu Q, Lu F. Effect of the Combined Compound Probiotics with Glycyrrhinic Acid on Alleviating Cytotoxicity of IPEC-J2 Cells Induced by Multi-Mycotoxins. Toxins (Basel) 2022; 14:toxins14100670. [PMID: 36287939 PMCID: PMC9612255 DOI: 10.3390/toxins14100670] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/17/2022] [Accepted: 09/22/2022] [Indexed: 11/17/2022] Open
Abstract
Aflatoxins B1 (AFB1), deoxynivalenol (DON) and zearalenone (ZEA) are the three most prevalent mycotoxins, whose contamination of food and feed is a severe worldwide problem. In order to alleviate the toxic effects of multi-mycotoxins (AFB1 + DON + ZEA, ADZ) on inflammation and apoptosis in swine jejunal epithelial cells (IPEC-J2), three species of probiotics (Bacillus subtilis, Saccharomyces cerevisiae and Pseudomonas lactis at 1 × 105 CFU/mL, respectively) were mixed together to make compound probiotics (CP), which were further combined with 400 μg/mL of glycyrrhinic acid (GA) to make bioactive materials (CGA). The experiment was divided into four groups, i.e., the control, ADZ, CGA and ADZ + CGA groups. The results showed that ADZ decreased cell viability and induced cytotoxicity, while CGA addition could alleviate ADZ-induced cytotoxicity. Moreover, the mRNA expressions of IL-8, TNF-α, NF-Κb, Bcl-2, Caspase-3, ZO-1, Occludin, Claudin-1 and ASCT2 genes, and protein expressions of TNF-α and Claudin-1 were significantly upregulated in ADZ group; while the mRNA abundances of IL-8, TNF-α, NF-Κb, Caspase-3, ASCT2 genes, and protein expressions of TNF-α and Claudin-1 were significantly downregulated in the ADZ + CGA group. In addition, the protein expressions of COX-2, ZO-1, and ASCT2 were significantly downregulated in the ADZ group, compared with the control group; whereas CGA co-incubation with ADZ could increase these protein expressions to recover to normal levels. This study indicated that CGA could alleviate cytotoxicity, apoptosis and inflammation in ADZ-induced IPEC-J2 cells and protect intestinal cell integrity from ADZ damages.
Collapse
Affiliation(s)
- Lijun Wang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Xiaomin Wang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Juan Chang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Ping Wang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Chaoqi Liu
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Lin Yuan
- Institute of Animal Husbandry and Veterinary Medicine, Henan Academy of Agricultural Sciences, Zhengzhou 450003, China
| | - Qingqiang Yin
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
- Correspondence:
| | - Qun Zhu
- Henan Delin Biological Product Co., Ltd., Xinxiang 453000, China
| | - Fushan Lu
- Henan Puai Feed Co., Ltd., Zhoukou 466000, China
| |
Collapse
|
37
|
Luo Z, Liu C, Hu Y, Xia T, Zhang B, Chen F, Tan X, Zheng Z. Gegen Qinlian decoction restores the intestinal barrier in bacterial diarrhea piglets by promoting Lactobacillus growth and inhibiting the TLR2/MyD88/NF-κB pathway. Biomed Pharmacother 2022; 155:113719. [PMID: 36152417 DOI: 10.1016/j.biopha.2022.113719] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 09/05/2022] [Accepted: 09/19/2022] [Indexed: 11/17/2022] Open
Abstract
Acute bacterial diarrhea is a severe global problem with a particularly high incidence rate in children. The microecology inhabiting the intestinal mucosa is the key factor leading to diarrhea. Gegen Qinlian decoction (GQD) is used to treat bacterial diarrhea, however, its underlying mechanism remains unclear. Thus, this study aimed to clarify the restorative effect of GQD on the intestinal barrier from the perspective of gut microbiota. A Tibetan piglet model with bacterial diarrhea was established through orally administered Escherichia coli, and diarrheal piglets were treated with GQD for three days. After treatment, GQD significantly ameliorated the diarrheal symptoms. GQD decreased the levels of IL-6, LPS, and DAO, and increased SIgA, ZO-1, and occludin levels in intestinal mucosa, indicating the restoration of intestinal barrier. GQD modulated the microbial compositions inhabited on the intestinal mucosa, especially an increase of the Lactobacillus. Spearman analysis showed that Lactobacillus was the key genus of intestinal barrier-related bacteria. Bacterial culture in vitro validated that GQD directly promoted Lactobacillus growth and inhibited E. coli proliferation. Moreover, the expressions of TLR2, MyD88, and NF-κB in the colon decreased after GQD treatment. In conclusion, GQD may treat diarrhea and restore the intestinal mucosal barrier by facilitating Lactobacillus growth and inhibiting the TLR2/MyD88/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Zhenye Luo
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou 510515, PR China; Guangdong Provincial Engineering Laboratory of Chinese Medicine Preparation Technology, Guangzhou 510515, PR China.
| | - Changshun Liu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou 510515, PR China; Guangdong Provincial Engineering Laboratory of Chinese Medicine Preparation Technology, Guangzhou 510515, PR China.
| | - Yannan Hu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou 510515, PR China; Guangdong Provincial Engineering Laboratory of Chinese Medicine Preparation Technology, Guangzhou 510515, PR China.
| | - Ting Xia
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou 510515, PR China; Guangdong Provincial Engineering Laboratory of Chinese Medicine Preparation Technology, Guangzhou 510515, PR China.
| | - Baoping Zhang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou 510515, PR China; Guangdong Provincial Engineering Laboratory of Chinese Medicine Preparation Technology, Guangzhou 510515, PR China.
| | - Feilong Chen
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou 510515, PR China; Guangdong Provincial Engineering Laboratory of Chinese Medicine Preparation Technology, Guangzhou 510515, PR China.
| | - Xiaomei Tan
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou 510515, PR China; Guangdong Provincial Engineering Laboratory of Chinese Medicine Preparation Technology, Guangzhou 510515, PR China.
| | - Zezhong Zheng
- South China Agricultural University College of Veterinary Medicine, Guangzhou 510642, PR China.
| |
Collapse
|
38
|
Yang Y, Fan L, Peng Y, Peng C, Li X. Alcohol–soluble polysaccharides from Dendrobium officinale flowers as an antidepressant by regulating the gut–brain axis. Int J Biol Macromol 2022; 216:836-849. [DOI: 10.1016/j.ijbiomac.2022.07.220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 07/14/2022] [Accepted: 07/27/2022] [Indexed: 11/05/2022]
|
39
|
Fang M, Hu W, Liu B. Protective and detoxifying effects conferred by selenium against mycotoxins and livestock viruses: A review. Front Vet Sci 2022; 9:956814. [PMID: 35982930 PMCID: PMC9378959 DOI: 10.3389/fvets.2022.956814] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 07/06/2022] [Indexed: 11/13/2022] Open
Abstract
Animal feed can easily be infected with molds during production and storage processes, and this can lead to the production of secondary metabolites, such as mycotoxins, which eventually threaten human and animal health. Furthermore, livestock production is also not free from viral infections. Under these conditions, the essential trace element, selenium (Se), can confer various biological benefits to humans and animals, especially due to its anticancer, antiviral, and antioxidant properties, as well as its ability to regulate immune responses. This article reviews the latest literature on the antagonistic effects of Se on mycotoxin toxicity and viral infections in animals. We outlined the systemic toxicity of mycotoxins and the primary mechanisms of mycotoxin-induced toxicity in this analysis. In addition, we pay close attention to how mycotoxins and viral infections in livestock interact. The use of Se supplementation against mycotoxin-induced toxicity and cattle viral infection was the topic of our final discussion. The coronavirus disease 2019 (COVID-19) pandemic, which is currently causing a health catastrophe, has altered our perspective on health concerns to one that is more holistic and increasingly embraces the One Health Concept, which acknowledges the interdependence of humans, animals, and the environment. In light of this, we have made an effort to present a thorough and wide-ranging background on the protective functions of selenium in successfully reducing mycotoxin toxicity and livestock viral infection. It concluded that mycotoxins could be systemically harmful and pose a severe risk to human and animal health. On the contrary, animal mycotoxins and viral illnesses have a close connection. Last but not least, these findings show that the interaction between Se status and host response to mycotoxins and cattle virus infection is crucial.
Collapse
Affiliation(s)
- Manxin Fang
- College of Life Science and Resources and Environment, Yichun University, Yichun, China
- Engineering Technology Research Center of Jiangxi Universities and Colleges for Selenium Agriculture, Yichun University, Yichun, China
- *Correspondence: Manxin Fang
| | - Wei Hu
- College of Life Science and Resources and Environment, Yichun University, Yichun, China
- Engineering Technology Research Center of Jiangxi Universities and Colleges for Selenium Agriculture, Yichun University, Yichun, China
| | - Ben Liu
- College of Life Science and Resources and Environment, Yichun University, Yichun, China
- Engineering Technology Research Center of Jiangxi Universities and Colleges for Selenium Agriculture, Yichun University, Yichun, China
| |
Collapse
|
40
|
Liu S, Kang W, Mao X, Du H, Ge L, Hou L, Yuan X, Wang M, Chen X, Liu Y, Huang K. Low dose of arsenic exacerbates toxicity to mice and IPEC-J2 cells exposed with deoxynivalenol: Aryl hydrocarbon receptor and autophagy might be novel therapeutic targets. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 832:155027. [PMID: 35381244 DOI: 10.1016/j.scitotenv.2022.155027] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 03/28/2022] [Accepted: 03/31/2022] [Indexed: 06/14/2023]
Abstract
Deoxynivalenol (DON) and arsenic (As) are widespread environmental contaminants, which are frequently found in human and animal food products. The intestine is a common target of As and DON when they are digested. Numerous studies mainly evaluate the individual effects whereas their combined toxicity has rarely been elucidated. Hence, this study was to assess the effect of low dose of NaAsO2 on DON-induced intestinal damage and explore the underling mechanism in mice and IPEC-J2 cells. The results showed that low dose of NaAsO2 exacerbated DON-induced intestinal impairment by increasing intestinal permeability and decreasing the abundance of tight junction proteins (ZO-1, Occludin, Claudin-1). Further, low dose of NaAsO2 enhanced the AhR signaling pathway and autophagy-related mRNA/protein expressions induced by DON. Interestingly, FICZ, an AhR activator, instead of CH223191, an AhR inhibitor, could alleviate toxicity of the low dose of NaAsO2 in the mice and IPEC-J2 cells. Compared to the WT IPEC-J2 cells, the intestinal barrier damage was more serious in LC3B-/- IPEC-J2 cells induced by low dose of NaAsO2 combination with DON. Collectively, our study demonstrated that low dose of NaAsO2 exacerbated DON-induced intestinal barrier impairment in vivo and in vitro. The present study also demonstrated that activation of AhR-mediated autophagy might be a self-protection mechanism. Hence, AhR and autophagy might be novel therapeutic targets to prevent or alleviate NaAsO2 combined with DON-induced intestinal barrier impairment.
Collapse
Affiliation(s)
- Shuiping Liu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China; Institute of Animal Nutritional Health, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China
| | - Weili Kang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China; Institute of Animal Nutritional Health, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China
| | - Xinru Mao
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China; Institute of Animal Nutritional Health, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China
| | - Heng Du
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China; Institute of Animal Nutritional Health, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China
| | - Lei Ge
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China; Institute of Animal Nutritional Health, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China
| | - Lili Hou
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China; Institute of Animal Nutritional Health, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China
| | - Xin Yuan
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China; Institute of Animal Nutritional Health, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China
| | - Mengmeng Wang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China; Institute of Animal Nutritional Health, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China
| | - Xingxiang Chen
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China; Institute of Animal Nutritional Health, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China
| | - Yunhuan Liu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China; Institute of Animal Nutritional Health, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China
| | - Kehe Huang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China; Institute of Animal Nutritional Health, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China.
| |
Collapse
|
41
|
Gou HZ, Zhang YL, Ren LF, Li ZJ, Zhang L. How do intestinal probiotics restore the intestinal barrier? Front Microbiol 2022; 13:929346. [PMID: 35910620 PMCID: PMC9330398 DOI: 10.3389/fmicb.2022.929346] [Citation(s) in RCA: 56] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 06/27/2022] [Indexed: 12/14/2022] Open
Abstract
The intestinal barrier is a structure that prevents harmful substances, such as bacteria and endotoxins, from penetrating the intestinal wall and entering human tissues, organs, and microcirculation. It can separate colonizing microbes from systemic tissues and prevent the invasion of pathogenic bacteria. Pathological conditions such as shock, trauma, stress, and inflammation damage the intestinal barrier to varying degrees, aggravating the primary disease. Intestinal probiotics are a type of active microorganisms beneficial to the health of the host and an essential element of human health. Reportedly, intestinal probiotics can affect the renewal of intestinal epithelial cells, and also make cell connections closer, increase the production of tight junction proteins and mucins, promote the development of the immune system, regulate the release of intestinal antimicrobial peptides, compete with pathogenic bacteria for nutrients and living space, and interact with the host and intestinal commensal flora to restore the intestinal barrier. In this review, we provide a comprehensive overview of how intestinal probiotics restore the intestinal barrier to provide new ideas for treating intestinal injury-related diseases.
Collapse
Affiliation(s)
- Hong-Zhong Gou
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, China
| | - Yu-Lin Zhang
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, China
| | - Long-Fei Ren
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, China
| | - Zhen-Jiao Li
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, China
| | - Lei Zhang
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, China
- *Correspondence: Lei Zhang,
| |
Collapse
|
42
|
An J, Liu Y, Wang Y, Fan R, Hu X, Zhang F, Yang J, Chen J. The Role of Intestinal Mucosal Barrier in Autoimmune Disease: A Potential Target. Front Immunol 2022; 13:871713. [PMID: 35844539 PMCID: PMC9284064 DOI: 10.3389/fimmu.2022.871713] [Citation(s) in RCA: 55] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Accepted: 06/08/2022] [Indexed: 11/13/2022] Open
Abstract
Autoimmune diseases are a series of diseases involving multiple tissues and organs, characterized by the over production of abnormal multiple antibodies. Although most studies support that the impaired immune balance participates in the development of autoimmune diseases, the specific pathogenesis of it is not fully understood. Intestinal immunity, especially the intestinal mucosal barrier has become a research hotspot, which is considered to be an upstream mechanism leading to the impaired immune balance. As an important defense barrier, the intestinal mucosal barrier regulates and maintains the homeostasis of internal environment. Once the intestinal barrier function is impaired under the effect of multiple factors, it will destroy the immune homeostasis, trigger inflammatory response, and participate in the development of autoimmune diseases in the final. However, the mechanism of the intestinal mucosal barrier how to regulate the homeostasis and inflammation is not clear. Some studies suggest that it maintains the balance of immune homeostasis through the zonulin pathway, intestinal microbiome, and Toll-like receptor signaling pathway. Our review focused on the composition and the function of the intestinal mucosal barrier to describe the research progress of it in regulating the immune homeostasis and inflammation, and also pointed that the intestinal mucosal barrier was the potential targets in the treatment of autoimmune diseases.
Collapse
Affiliation(s)
- Jia An
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Yuqing Liu
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Yiqi Wang
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Ru Fan
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Xiaorong Hu
- Department of Microbiology and Immunology, Shanxi Medical University, Taiyuan, China
| | - Fen Zhang
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Jinhua Yang
- Department of Internal Medicine, Central Hospital of Xinghualing District, Taiyuan, China
| | - Junwei Chen
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, China
- *Correspondence: Junwei Chen,
| |
Collapse
|
43
|
Hao W, Li A, Wang J, An G, Guan S. Mycotoxin Contamination of Feeds and Raw Materials in China in Year 2021. Front Vet Sci 2022; 9:929904. [PMID: 35847652 PMCID: PMC9281542 DOI: 10.3389/fvets.2022.929904] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 06/01/2022] [Indexed: 11/15/2022] Open
Abstract
In this research, we performed a large-scale survey of mycotoxin contamination in several feed commodities and assessed regional differences in mycotoxin occurrence in maize across China in 2021. Concentrations of aflatoxins, zearalenone (ZEN), fumonisins, and trichothecenes type B were analyzed in 2,643 raw material and compound feed samples collected from eight provinces. Generally, trichothecenes type B, fumonisins, and ZEN were most prevalent and detected in averages of positive concentrations at 1,167, 1,623, and 204 μg/kg, respectively. In the new season maize, samples were also seriously infested with trichothecenes type B, fumonisins, and ZEN, and their averages of positive concentrations were 1,302, 2,518, and 225 μg/kg, respectively. Wheat was commonly contaminated with trichothecenes type B and ZEN, and the highest concentration levels of trichothecenes type B, fumonisins, and ZEN were all detected in the samples from maize by-products. Among the different geographical regions, distinct trends were observed in new season maize. Samples from Shandong province were highly contaminated with trichothecenes type B, fumonisins, and ZEN, while special attention should be paid to aflatoxins and fumonisins in Anhui and Jiangsu provinces in East China. In addition, the present survey showed that compound feeds and raw materials are commonly contaminated by multiple mycotoxins. Trichothecenes type B and ZEN concentrations were correlated significantly in this survey.
Collapse
Affiliation(s)
- Wei Hao
- Department of Animal Nutrition and Health, DSM (China) Co., Ltd, Shanghai, China
| | - Anping Li
- Department of Animal Nutrition and Health, DSM (China) Co., Ltd, Shanghai, China
| | - Jinyong Wang
- Department of Animal Nutrition and Health, DSM (China) Co., Ltd, Shanghai, China
| | - Gang An
- Department of Animal Nutrition and Health, DSM (China) Co., Ltd, Shanghai, China
| | - Shu Guan
- Department of Animal Nutrition and Health, DSM Singapore Industrial Pte Ltd, Singapore, Singapore
- *Correspondence: Shu Guan
| |
Collapse
|
44
|
Bai J, Zhou Y, Luo X, Hai J, Si X, Li J, Fu H, Dai Z, Yang Y, Wu Z. Roles of stress response-related signaling and its contribution to the toxicity of zearalenone in mammals. Compr Rev Food Sci Food Saf 2022; 21:3326-3345. [PMID: 35751400 DOI: 10.1111/1541-4337.12974] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 04/06/2022] [Accepted: 04/11/2022] [Indexed: 11/30/2022]
Abstract
Zearalenone (ZEA) is a mycotoxin frequently found in cereal crops and cereal-derived foodstuffs worldwide. It affects plant productivity, and is also a serious hazard to humans and animals if being exposed to food/feed contaminated by ZEA. Studies over the last decade have shown that the toxicity of ZEA in animals is mainly mediated by the various stress responses, such as endoplasmic reticulum (ER) stress, oxidative stress, and others. Accumulating evidence shows that oxidative stress and ER stress signaling are actively implicated in and contributes to the pathophysiology of various diseases. Biochemically, the deleterious effects of ZEA are associated with apoptosis, DNA damage, and lipid peroxidation by regulating the expression of genes implicated in these biological processes. Despite these findings, the underlying mechanisms responsible for these alterations remain unclear. This review summarized the characteristics, metabolism, toxicity and the deleterious effects of ZEA exposure in various tissues of animals. Stress response signaling implicated in the toxicity as well as potential therapeutic options with the ability to reduce the deleterious effects of ZEA in animals were highlighted and discussed.
Collapse
Affiliation(s)
- Jun Bai
- State Key Laboratory of Animal Nutrition, Department of Companion Animal Science, College of Animal Science and Technology, China Agricultural University, Beijing, P. R. China
| | - Yusong Zhou
- State Key Laboratory of Animal Nutrition, Department of Companion Animal Science, College of Animal Science and Technology, China Agricultural University, Beijing, P. R. China
| | - Xin Luo
- State Key Laboratory of Animal Nutrition, Department of Companion Animal Science, College of Animal Science and Technology, China Agricultural University, Beijing, P. R. China
| | - Jia Hai
- State Key Laboratory of Animal Nutrition, Department of Companion Animal Science, College of Animal Science and Technology, China Agricultural University, Beijing, P. R. China
| | - Xuemeng Si
- State Key Laboratory of Animal Nutrition, Department of Companion Animal Science, College of Animal Science and Technology, China Agricultural University, Beijing, P. R. China
| | - Jun Li
- State Key Laboratory of Animal Nutrition, Department of Companion Animal Science, College of Animal Science and Technology, China Agricultural University, Beijing, P. R. China
| | - Huiyang Fu
- State Key Laboratory of Animal Nutrition, Department of Companion Animal Science, College of Animal Science and Technology, China Agricultural University, Beijing, P. R. China
| | - Zhaolai Dai
- State Key Laboratory of Animal Nutrition, Department of Companion Animal Science, College of Animal Science and Technology, China Agricultural University, Beijing, P. R. China
| | - Ying Yang
- State Key Laboratory of Animal Nutrition, Department of Companion Animal Science, College of Animal Science and Technology, China Agricultural University, Beijing, P. R. China
| | - Zhenlong Wu
- State Key Laboratory of Animal Nutrition, Department of Companion Animal Science, College of Animal Science and Technology, China Agricultural University, Beijing, P. R. China.,Beijing Jingwa Agricultural Science and Technology Innovation Center, #1, Yuda Road, Pinggu, Beijing, P. R. China
| |
Collapse
|
45
|
Tao Y, Qiao S, Lv C, Yun X, Yue M, Fang Y, Wei Z, Dai Y, Xia Y. Phytoestrogen arctigenin preserves the mucus barrier in inflammatory bowel diseases by inhibiting goblet cell apoptosis via the
ERβ
/
TRIM21
/
PHB1
pathway. Phytother Res 2022; 36:3248-3264. [DOI: 10.1002/ptr.7495] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 04/26/2022] [Accepted: 04/30/2022] [Indexed: 01/17/2023]
Affiliation(s)
- Yu Tao
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy China Pharmaceutical University Nanjing China
- Jiangsu Province Hospital of Chinese Medicine The Affiliated Hospital of Nanjing University of Chinese Medicine Nanjing China
| | - Si‐miao Qiao
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy China Pharmaceutical University Nanjing China
| | - Chang‐jun Lv
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy China Pharmaceutical University Nanjing China
| | - Xin‐ming Yun
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy China Pharmaceutical University Nanjing China
| | - Meng‐fan Yue
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy China Pharmaceutical University Nanjing China
| | - Yu‐lai Fang
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy China Pharmaceutical University Nanjing China
| | - Zhi‐feng Wei
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy China Pharmaceutical University Nanjing China
| | - Yue Dai
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy China Pharmaceutical University Nanjing China
| | - Yu‐feng Xia
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy China Pharmaceutical University Nanjing China
| |
Collapse
|
46
|
Li G, Jin B, Fan Z. Mechanisms Involved in Gut Microbiota Regulation of Skeletal Muscle. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:2151191. [PMID: 35633886 PMCID: PMC9132697 DOI: 10.1155/2022/2151191] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 05/03/2022] [Indexed: 12/12/2022]
Abstract
Skeletal muscle is one of the largest organs in the body and is essential for maintaining quality of life. Loss of skeletal muscle mass and function can lead to a range of adverse consequences. The gut microbiota can interact with skeletal muscle by regulating a variety of processes that affect host physiology, including inflammatory immunity, protein anabolism, energy, lipids, neuromuscular connectivity, oxidative stress, mitochondrial function, and endocrine and insulin resistance. It is proposed that the gut microbiota plays a role in the direction of skeletal muscle mass and work. Even though the notion of the gut microbiota-muscle axis (gut-muscle axis) has been postulated, its causal link is still unknown. The impact of the gut microbiota on skeletal muscle function and quality is described in detail in this review.
Collapse
Affiliation(s)
- Guangyao Li
- Department of General Surgery, The Third People's Hospital of Dalian, Dalian Medical University, Dalian, China
- Department of Central Laboratory, The Third People's Hospital of Dalian, Dalian Medical University, Dalian, China
| | - Binghui Jin
- Department of General Surgery, The Third People's Hospital of Dalian, Dalian Medical University, Dalian, China
- Department of Central Laboratory, The Third People's Hospital of Dalian, Dalian Medical University, Dalian, China
| | - Zhe Fan
- Department of General Surgery, The Third People's Hospital of Dalian, Dalian Medical University, Dalian, China
- Department of Central Laboratory, The Third People's Hospital of Dalian, Dalian Medical University, Dalian, China
| |
Collapse
|
47
|
Fathima S, Shanmugasundaram R, Adams D, Selvaraj RK. Gastrointestinal Microbiota and Their Manipulation for Improved Growth and Performance in Chickens. Foods 2022; 11:1401. [PMID: 35626971 PMCID: PMC9140538 DOI: 10.3390/foods11101401] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Revised: 05/01/2022] [Accepted: 05/09/2022] [Indexed: 12/17/2022] Open
Abstract
The gut of warm-blooded animals is colonized by microbes possibly constituting at least 100 times more genetic material of microbial cells than that of the somatic cells of the host. These microbes have a profound effect on several physiological functions ranging from energy metabolism to the immune response of the host, particularly those associated with the gut immune system. The gut of a newly hatched chick is typically sterile but is rapidly colonized by microbes in the environment, undergoing cycles of development. Several factors such as diet, region of the gastrointestinal tract, housing, environment, and genetics can influence the microbial composition of an individual bird and can confer a distinctive microbiome signature to the individual bird. The microbial composition can be modified by the supplementation of probiotics, prebiotics, or synbiotics. Supplementing these additives can prevent dysbiosis caused by stress factors such as infection, heat stress, and toxins that cause dysbiosis. The mechanism of action and beneficial effects of probiotics vary depending on the strains used. However, it is difficult to establish a relationship between the gut microbiome and host health and productivity due to high variability between flocks due to environmental, nutritional, and host factors. This review compiles information on the gut microbiota, dysbiosis, and additives such as probiotics, postbiotics, prebiotics, and synbiotics, which are capable of modifying gut microbiota and elaborates on the interaction of these additives with chicken gut commensals, immune system, and their consequent effects on health and productivity. Factors to be considered and the unexplored potential of genetic engineering of poultry probiotics in addressing public health concerns and zoonosis associated with the poultry industry are discussed.
Collapse
Affiliation(s)
- Shahna Fathima
- Department of Poultry Science, The University of Georgia, Athens, GA 30605, USA; (S.F.); (D.A.); (R.K.S.)
| | - Revathi Shanmugasundaram
- Toxicology and Mycotoxin Research Unit, US National Poultry Research Center, Athens, GA 30605, USA
| | - Daniel Adams
- Department of Poultry Science, The University of Georgia, Athens, GA 30605, USA; (S.F.); (D.A.); (R.K.S.)
| | - Ramesh K. Selvaraj
- Department of Poultry Science, The University of Georgia, Athens, GA 30605, USA; (S.F.); (D.A.); (R.K.S.)
| |
Collapse
|
48
|
Ma H, Li X, Yang H, Qiu Y, Xiao W. The Pathology and Physiology of Ileostomy. Front Nutr 2022; 9:842198. [PMID: 35529469 PMCID: PMC9072868 DOI: 10.3389/fnut.2022.842198] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 03/30/2022] [Indexed: 12/02/2022] Open
Abstract
An ileostomy is a surgery that is commonly performed to protect low pelvic anastomoses or prevent high-risk anastomotic leakages. However, various postoperative complications remain of major concern. After an ileostomy, the distal intestinal segment is left open for an extended period and is in a non-functional state. Consequently, the intestinal mucosa, smooth muscle, and microbiota undergo significant changes that are closely related to postoperative recovery and complications. A systematic description of these changes is necessary to understand the relationship among them and take more effective measures for postoperative intervention.
Collapse
Affiliation(s)
- Haitao Ma
- Department of General Surgery, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Xiaolong Li
- Department of General Surgery, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Hua Yang
- Department of General Surgery, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Yuan Qiu
- Department of General Surgery, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Weidong Xiao
- Department of General Surgery, Xinqiao Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
49
|
Zhao Q, Huang M, Yin J, Wan Y, Liu Y, Duan R, Luo Y, Xu X, Cao X, Yi M. Atrazine exposure and recovery alter the intestinal structure, bacterial composition and intestinal metabolites of male Pelophylax nigromaculatus. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 818:151701. [PMID: 34798088 DOI: 10.1016/j.scitotenv.2021.151701] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 11/11/2021] [Accepted: 11/11/2021] [Indexed: 06/13/2023]
Abstract
The pesticide atrazine poses a potential threat to the health of frogs living in farmland areas. The exposure concentration in traditional pesticide experiments is usually constant, while pesticide pollution in actual water may fluctuate due to periodic or seasonal application. We examined the effects of different concentrations of atrazine (50, 100 and 500 μg/L) over a 14-day exposure and a 7-day recovery on intestinal histology, bacterial composition and intestinal metabolites of male Pelophylax nigromaculatus. HE staining revealed that after a 14-day atrazine exposure, the 100 μg/L and 500 μg/L groups showed obvious cysts and significantly decreased intestinal crypt depth and villus height. After a 7-day recovery, the damaged intestine in the 100 μg/L group was partially recovered, while in the 500 μg/L exposure group there was no improvement. 16S rRNA gene analysis of intestinal bacteria showed that 500 μg/L atrazine exposure significantly caused a persistent decrease in bacterial α diversity. Compared to the control and other atrazine exposure groups, the 500 μg/L group showed significant changes in the relative abundance of predominant bacteria. In addition, most dominant bacteria in the 500 μg/L recovery group showed significant differences with the 50 μg/L and 100 μg/L recovery groups. Nontargeted metabolomics profiling based on UPLC/MS analysis showed that atrazine exposure and recovery induced changes in the intestinal metabolic profile. The changes in metabolites were mainly related to purine/pyrimidine metabolism, glycine, serine and threonine metabolism, and arginine and proline metabolism. In general, these pathways were closely related to energy metabolism and amino acid metabolism. These results suggest that the short-term exposure to 500 μg/L atrazine causes persistent harm to intestinal health. This study is an important step toward a better understanding of the toxic effects of atrazine exposure and recovery in frog intestines.
Collapse
Affiliation(s)
- Qiang Zhao
- College of Agriculture and Biotechnology, Hunan University of Humanities, Science and Technology, Loudi 417000, Hunan, China
| | - Minyi Huang
- College of Agriculture and Biotechnology, Hunan University of Humanities, Science and Technology, Loudi 417000, Hunan, China.
| | - Jiawei Yin
- College of Agriculture and Biotechnology, Hunan University of Humanities, Science and Technology, Loudi 417000, Hunan, China
| | - Yuyue Wan
- College of Agriculture and Biotechnology, Hunan University of Humanities, Science and Technology, Loudi 417000, Hunan, China
| | - Yang Liu
- College of Agriculture and Biotechnology, Hunan University of Humanities, Science and Technology, Loudi 417000, Hunan, China
| | - Renyan Duan
- College of Agriculture and Biotechnology, Hunan University of Humanities, Science and Technology, Loudi 417000, Hunan, China
| | - Yucai Luo
- College of Agriculture and Biotechnology, Hunan University of Humanities, Science and Technology, Loudi 417000, Hunan, China
| | - Xiang Xu
- College of Agriculture and Biotechnology, Hunan University of Humanities, Science and Technology, Loudi 417000, Hunan, China
| | - Xiaohong Cao
- College of Agriculture and Biotechnology, Hunan University of Humanities, Science and Technology, Loudi 417000, Hunan, China
| | - Minghui Yi
- College of Agriculture and Biotechnology, Hunan University of Humanities, Science and Technology, Loudi 417000, Hunan, China
| |
Collapse
|
50
|
Chang L, Ding Y, Wang Y, Song Z, Li F, He X, Zhang H. Effects of Different Oligosaccharides on Growth Performance and Intestinal Function in Broilers. Front Vet Sci 2022; 9:852545. [PMID: 35433897 PMCID: PMC9011052 DOI: 10.3389/fvets.2022.852545] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 03/01/2022] [Indexed: 11/13/2022] Open
Abstract
ObjectiveThis study was conducted to investigate the effects of different oligosaccharides on the growth performance and intestinal function in broilers.MethodsA total of 360 1-day-old yellow-feather chickens were randomly divided into 5 groups and fed with a basal diet supplemented with 50 mg/kg chlortetracycline (ANT), 3 g/kg isomalto-oligosaccharide (IMO), 3 g/kg raffinose oligosaccharide (RFO), and 30 mg/kg chitooligosaccharide (COS). The experiment lasted for 56 days, with 1–28 days as the starter phase and 29–56 days as the grower phase.ResultsThe results showed that dietary supplementation with RFO and COS significantly improved average daily gain (ADG) and average daily feed intake (ADFI) (p < 0.05). Relative to the control group, diets supplemented with oligosaccharides dramatically increased the level of serum IgM (RFO, COS), T-SOD (COS), and GSH-Px (IMO and RFO) and the expression of ZO-1(IMO) and claudin-1 (RFO) (p < 0.05). Adding antibiotics or oligosaccharides to the diet could remarkedly increase the villus height and villus height (VH)/crypt depth (CD) ratio of each group (p < 0.05). Through the ileum α-diversity analysis and comparison of OTU number in each group showed that the microbial richness of the IMO group increased in the starter phase, and that of the RFO and CSO group increased in the grower phase. Additionally, compared with the control group, IMO supplementation increased the level of ileum sIgA (p < 0.05) and the content of valeric acid (p < 0.05) in the cecum.ConclusionsIn summary, the addition of oligosaccharides in diet can improve the immune function and antioxidant capacity and improve intestinal health of broilers.
Collapse
Affiliation(s)
- Ling Chang
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
- Provincial and Ministerial Co-construction of Collaborative Innovation Center for High-quality Animal Products Production, Changsha, China
- Hunan Engineering Research Center of Poultry Production Safety, Changsha, China
| | - Yanan Ding
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
- Provincial and Ministerial Co-construction of Collaborative Innovation Center for High-quality Animal Products Production, Changsha, China
- Hunan Engineering Research Center of Poultry Production Safety, Changsha, China
| | - Yushi Wang
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
- Provincial and Ministerial Co-construction of Collaborative Innovation Center for High-quality Animal Products Production, Changsha, China
- Hunan Engineering Research Center of Poultry Production Safety, Changsha, China
| | - Zehe Song
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
- Provincial and Ministerial Co-construction of Collaborative Innovation Center for High-quality Animal Products Production, Changsha, China
- Hunan Engineering Research Center of Poultry Production Safety, Changsha, China
| | - Fei Li
- Guangxi Fufeng Agriculture and Animal Husbandry Group Co., Ltd., Nanning, China
| | - Xi He
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
- Provincial and Ministerial Co-construction of Collaborative Innovation Center for High-quality Animal Products Production, Changsha, China
- Hunan Engineering Research Center of Poultry Production Safety, Changsha, China
| | - Haihan Zhang
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
- Provincial and Ministerial Co-construction of Collaborative Innovation Center for High-quality Animal Products Production, Changsha, China
- Hunan Engineering Research Center of Poultry Production Safety, Changsha, China
| |
Collapse
|