1
|
Shi Y, Liu J, Zhou H, Wu Z, Qiu Y, Ye C. Dihydromyricetin alleviates ETEC K88-induced intestinal inflammatory injury by inhibiting quorum sensing-related virulence factors. BMC Microbiol 2025; 25:201. [PMID: 40205366 PMCID: PMC11980137 DOI: 10.1186/s12866-025-03879-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 03/10/2025] [Indexed: 04/11/2025] Open
Abstract
BACKGROUND Enterotoxigenic Escherichia coli (ETEC) is responsible for piglet diarrhea and causes substantial economic loss in the pig industry. Along with the restriction of antibiotics, natural compounds targeting bacterial virulence factors are supposed to be efficacious and attractive alternatives for controlling ETEC infection. This study aimed to investigate the influence of dihydromyricetin (DMY), a natural flavonoid compound, on the expression of virulence factors of ETEC and intestinal inflammatory injury. RESULTS DMY interfered with the quorum sensing (QS) of ETEC K88 since it decreased AI-2 secretion and downregulated the expression of LuxS and Pfs, which dominate AI-2 production, and decreased the expression mRNA level of genes (lsrA, lsrB, lsrC, lsrD, lsrK, and lsrR) that are involved in AI-2 internalization and signal transduction. Additionally, DMY markedly dampened the expression of QS-related virulence genes (elt-1, estB, fliC, faeG), biofilm formation, cell adhesion, and stress tolerance of ETEC K88. Furthermore, DMY treatment applied to the ETEC K88 infection in mice model resulted in decreased amount of heat-labile (LT) and heat-stable (ST) enterotoxins, reduced production of cAMP and cGMP, downregulated protein level of CFTR and upregulated expression of NHE3 in the ileum. In addition, the mRNA expression of proinflammatory cytokines (TNF-α, IL-1β, and IL-6) and histological damage in the ileum were significantly decreased by DMY treatment. CONCLUSIONS DMY can inhibit the AI-2 QS and virulence factor expression, thereby attenuating the virulence of ETEC and alleviating intestinal inflammatory damage in ETEC K88-challenged mice. This study indicated that DMY has the potential to be a promising antivirulence agent for combating ETEC infection.
Collapse
Affiliation(s)
- Yaqian Shi
- Hubei Key Laboratory of Animal Nutrition and Feed Science, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan, China
- Wuhan Engineering and Technology Research Center of Animal Disease-resistant Nutrition, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan, China
| | - Jin Liu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan, China
- Wuhan Engineering and Technology Research Center of Animal Disease-resistant Nutrition, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan, China
| | - Hualin Zhou
- Agricultural College, Xiangyang Polytechnic, Xiangyang, China
| | - Zhongyuan Wu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan, China.
- Wuhan Engineering and Technology Research Center of Animal Disease-resistant Nutrition, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan, China.
| | - Yinsheng Qiu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan, China
- Wuhan Engineering and Technology Research Center of Animal Disease-resistant Nutrition, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan, China
| | - Chun Ye
- Hubei Key Laboratory of Animal Nutrition and Feed Science, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan, China
- Wuhan Engineering and Technology Research Center of Animal Disease-resistant Nutrition, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan, China
| |
Collapse
|
2
|
Demir R, Sarıtaş S, Bechelany M, Karav S. Lactoferrin: Properties and Potential Uses in the Food Industry. Int J Mol Sci 2025; 26:1404. [PMID: 40003872 PMCID: PMC11855648 DOI: 10.3390/ijms26041404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 02/01/2025] [Accepted: 02/04/2025] [Indexed: 02/27/2025] Open
Abstract
Lactoferrin (LF) is an 80 kDa glycoprotein that contains approximately 700 amino acids and is a member of the transferrin family. The essential properties of LF, including antimicrobial, antiviral, anticancer, anti-inflammatory, antioxidant, and probiotic effects, have been studied for decades. The iron chelation activity of LF is significantly associated with its antimicrobial, anti-inflammatory, and antioxidant properties. Owing to its probiotic and prebiotic activity, LF also facilitates the growth of beneficial microorganisms and iron-defense immediate-effect properties on pathogens. Additionally, the ability to regulate cell signaling pathways and immune responses makes LF a prominent modulatory protein. These diverse characteristics of LF have gained interest in its therapeutic potential. Studies have suggested that LF could serve as an alternative source to antibiotics in severe infections and illnesses. LF has also gained interest in the food industry for its potential as an additive to fortify products such as yogurt, infant formula, and meat derivatives while also improving the shelf life of foods and providing antimicrobial and antioxidant activity. Prior to using LF in the food industry, the safety and toxicity of food processing are necessary to be investigated. These safety investigations are crucial for addressing potential harm or side effects and ensuring a healthy lifestyle. This review discusses the attributes and safety of LF, particularly its exploitation in the food industry.
Collapse
Affiliation(s)
- Ranya Demir
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale 17000, Türkiye; (R.D.); (S.S.)
| | - Sümeyye Sarıtaş
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale 17000, Türkiye; (R.D.); (S.S.)
| | - Mikhael Bechelany
- Institut Européen des Membranes (IEM), UMR 5635, University Montpellier, ENSCM, CNRS, F-34095 Montpellier, France
- Functional Materials Group, Gulf University for Science and Technology (GUST), Masjid Al Aqsa Street, Mubarak Al-Abdullah 32093, Kuwait
| | - Sercan Karav
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale 17000, Türkiye; (R.D.); (S.S.)
| |
Collapse
|
3
|
Pramudito TE, Klostermann C, Smid EJ, Schols HA. Modulation of soy flour bioactivity against enterotoxigenic Escherichia coli by fermentation with exopolysaccharides-producing lactic acid bacteria. Carbohydr Polym 2025; 348:122922. [PMID: 39567144 DOI: 10.1016/j.carbpol.2024.122922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 10/08/2024] [Accepted: 10/25/2024] [Indexed: 11/22/2024]
Abstract
Enterotoxigenic Escherichia coli (ETEC)-mediated diarrhea can be mitigated by inhibiting bacterial adhesion to intestinal surface. Some lactic acid bacteria (LAB) produce exopolysaccharides (EPS) that can inhibit ETEC adhesion. In this study, we fermented soy flour-based dough (SoyD) with EPS-producing LAB strains Pediococcus pentosaceus TL (PpTL), Leuconostoc citreum TR (LcTR), Leuconostoc mesenteroides WA (LmWA) and L. mesenteroides WN (LmWN) to improve anti-adhesive activity of the dough against ETEC. The strains LcTR, LmWA and LmWN produced EPS in SoyD fermentation with similar polysaccharide yields and compositions as when grown in liquid medium, whereas PpTL was unable to produce EPS in SoyD. LcTR produced high molecular weight (Mw) dextran (∼900 kDa) while LmWA and LmWN produced dextran and levan with diverse Mw (∼20-1000 kDa). SoyD fermentation by EPS-producing LAB increased the capability of the SoyD extracts to adhere to ETEC cells and block ETEC adhesion to porcine mucin. After Mw-based fractionation, all extract-fractions (>3 kDA) of LmWA- and LmWN-fermented SoyD retained their blocking activity indicating that various Mw populations of the EPS contributes to bioactivity against ETEC. This study shows the potential of EPS-producing LAB strains as fermenting microorganisms in the development of a functional food product with anti-diarrheal properties.
Collapse
Affiliation(s)
- Theodorus Eko Pramudito
- Laboratory of Food Chemistry, Wageningen University & Research, P.O. Box 17, 6700 AA Wageningen, the Netherlands; Faculty of Biotechnology, Atma Jaya Catholic University of Indonesia, Indonesia
| | - Cynthia Klostermann
- Laboratory of Food Chemistry, Wageningen University & Research, P.O. Box 17, 6700 AA Wageningen, the Netherlands
| | - Eddy J Smid
- Food Microbiology, Wageningen University & Research, the Netherlands
| | - Henk A Schols
- Laboratory of Food Chemistry, Wageningen University & Research, P.O. Box 17, 6700 AA Wageningen, the Netherlands.
| |
Collapse
|
4
|
Dell’Anno M, Frazzini S, Reggi S, Ferri I, Scaglia E, Schiasselloni L, Inglesi A, Riva F, Verdile N, Pasquariello R, Rossi L. Evaluation of dietary supplementation of Ascophyllum nodosum and Lithothamnium calcareum as functional algae in F4+ Escherichia coli challenged piglets. Front Vet Sci 2024; 11:1430347. [PMID: 39309030 PMCID: PMC11412951 DOI: 10.3389/fvets.2024.1430347] [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: 05/09/2024] [Accepted: 08/07/2024] [Indexed: 09/25/2024] Open
Abstract
Introduction Despite progress in reducing antimicrobial use in the veterinary field, it is crucial to find alternatives to preserve effectiveness and limit antimicrobial resistance. In pig farming, pathogenic strains of E. coli are the main cause of gastrointestinal disorders and antibiotic use. In this field, algae represent an innovation in animal nutrition that aligns with livestock sustainability principles and provide a high content of functional molecules. Aim The aim of this study was to evaluate the impact of an innovative dietary combination of Ascophyllum nodosum and Lithothamnium calcareum, on growth, duodenum gene expression, jejunum intestinal morphology, and serum oxidative status in F4+ Escherichia coli challenged piglets. Materials and methods Forty-eight weaned pigs, aged 28 ± 2 days, were divided into two groups (n = 24 pigs/group): the control group was fed a commercial diet (CTRL), while the seaweeds group was fed a commercial diet supplemented with 1.5% A. nodosum and 0.5% L. calcareum for 27 days (ALGAE). After 13 days, 50% of animals in each group were challenged with a single dose of 108 CFU/dose of E. coli F4+, resulting in two infected groups (CTRL+ and ALGAE+, n = 12 pigs/group). Growth performance was assessed by measuring the individual body weight. At day 27, from six animals/group duodenum and jejunum sections were sampled for gene expression analysis via qRT-PCR and histological evaluation. Results and discussion The results indicated a significantly higher body weight in the ALGAE+ group compared to CTRL+ after 7 days post-challenge (p < 0.0001). Jejunum morphology revealed lower villus height, villus width and villus height/crypt depth ratio in CTRL+ compared to ALGAE+ (p < 0.05) suggesting a protective effect of seaweeds on gut health. Conclusion In conclusion, algae mixture exerted a protective effect against intestinal damage from E. coli F4+ infection proposing A. nodosum and L. calcareum supplementation as interesting strategy to support animal growth, enhance health and reduce antibiotic treatments in weaned piglets.
Collapse
Affiliation(s)
- Matteo Dell’Anno
- Department of Veterinary Medicine and Animal Sciences—DIVAS, University of Milan, Lodi, Italy
| | - Sara Frazzini
- Department of Veterinary Medicine and Animal Sciences—DIVAS, University of Milan, Lodi, Italy
| | - Serena Reggi
- Department of Veterinary Medicine and Animal Sciences—DIVAS, University of Milan, Lodi, Italy
| | - Irene Ferri
- Department of Veterinary Medicine and Animal Sciences—DIVAS, University of Milan, Lodi, Italy
| | - Elena Scaglia
- Department of Veterinary Medicine and Animal Sciences—DIVAS, University of Milan, Lodi, Italy
- Department Civil, Environmental, Architectural Engineering and Mathematics—DICATAM, University of Brescia, Brescia, Italy
| | - Linda Schiasselloni
- Department of Veterinary Medicine and Animal Sciences—DIVAS, University of Milan, Lodi, Italy
| | - Alessia Inglesi
- Department of Veterinary Medicine and Animal Sciences—DIVAS, University of Milan, Lodi, Italy
| | - Federica Riva
- Department of Veterinary Medicine and Animal Sciences—DIVAS, University of Milan, Lodi, Italy
| | - Nicole Verdile
- Department of Veterinary Medicine and Animal Sciences—DIVAS, University of Milan, Lodi, Italy
| | - Rolando Pasquariello
- Department of Agricultural and Environmental Sciences—Production, Landscape, Agroenergy–DISAA, University of Milan, Milan, Italy
| | - Luciana Rossi
- Department of Veterinary Medicine and Animal Sciences—DIVAS, University of Milan, Lodi, Italy
| |
Collapse
|
5
|
van Niekerk AA, Maluck S, Mag P, Kővágó C, Kerek Á, Jerzsele Á, Steinmetzer T, Pászti-Gere E. Antiviral Drug Candidate Repositioning for Streptococcus suis Infection in Non-Tumorigenic Cell Models. Biomedicines 2024; 12:783. [PMID: 38672139 PMCID: PMC11048155 DOI: 10.3390/biomedicines12040783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 03/28/2024] [Accepted: 03/29/2024] [Indexed: 04/28/2024] Open
Abstract
The increasing prevalence of antimicrobial resistance against zoonotic bacteria, including Streptococcus (S.) suis, highlights the need for new therapeutical strategies, including the repositioning of drugs. In this study, susceptibilities of bacterial isolates were tested toward ten different 3-amidinophenyalanine (Phe(3-Am)) derivatives via determination of minimum inhibitory concentration (MIC) values. Some of these protease inhibitors, like compounds MI-432, MI-471, and MI-476, showed excellent antibacterial effects against S. suis. Their drug interaction potential was investigated using human liver microsomal cytochrome P450 (CYP450) measurements. In our work, non-tumorigenic IPEC-J2 cells and primary porcine hepatocytes were infected with S. suis, and the putative beneficial impact of these inhibitors was investigated on cell viability (Neutral red assay), on interleukin (IL)-6 levels (ELISA technique), and on redox balance (Amplex red method). The antibacterial inhibitors prevented S. suis-induced cell death (except MI-432) and decreased proinflammatory IL-6 levels. It was also found that MI-432 and MI-476 had antioxidant effects in an intestinal cell model upon S. suis infection. Concentration-dependent suppression of CYP3A4 function was found via application of all three inhibitors. In conclusion, our study suggests that the potential antiviral Phe(3-Am) derivatives with 2',4' dichloro-biphenyl moieties can be considered as effective drug candidates against S. suis infection due to their antibacterial effects.
Collapse
Affiliation(s)
- Ashley Anzet van Niekerk
- Department of Pharmacology and Toxicology, University of Veterinary Medicine, István utca 2, H-1078 Budapest, Hungary (Á.J.)
| | - Sara Maluck
- Department of Pharmacology and Toxicology, University of Veterinary Medicine, István utca 2, H-1078 Budapest, Hungary (Á.J.)
| | - Patrik Mag
- Department of Pharmacology and Toxicology, University of Veterinary Medicine, István utca 2, H-1078 Budapest, Hungary (Á.J.)
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine, István utca 2, H-1078 Budapest, Hungary
| | - Csaba Kővágó
- Department of Pharmacology and Toxicology, University of Veterinary Medicine, István utca 2, H-1078 Budapest, Hungary (Á.J.)
| | - Ádám Kerek
- Department of Pharmacology and Toxicology, University of Veterinary Medicine, István utca 2, H-1078 Budapest, Hungary (Á.J.)
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine, István utca 2, H-1078 Budapest, Hungary
| | - Ákos Jerzsele
- Department of Pharmacology and Toxicology, University of Veterinary Medicine, István utca 2, H-1078 Budapest, Hungary (Á.J.)
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine, István utca 2, H-1078 Budapest, Hungary
| | - Torsten Steinmetzer
- Faculty of Pharmacy, Institute of Pharmaceutical Chemistry, Philipps University Marburg, Marbacher Weg 6, 35032 Marburg, Germany
| | - Erzsébet Pászti-Gere
- Department of Pharmacology and Toxicology, University of Veterinary Medicine, István utca 2, H-1078 Budapest, Hungary (Á.J.)
| |
Collapse
|
6
|
Pramudito TE, Desai K, Voigt C, Smid EJ, Schols HA. Dextran and levan exopolysaccharides from tempeh-associated lactic acid bacteria with bioactivity against enterotoxigenic Escherichia coli (ETEC). Carbohydr Polym 2024; 328:121700. [PMID: 38220337 DOI: 10.1016/j.carbpol.2023.121700] [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: 12/12/2023] [Accepted: 12/13/2023] [Indexed: 01/16/2024]
Abstract
Soybean tempeh contains bioactive carbohydrate that can reduce the severity of diarrhea by inhibiting enterotoxigenic Escherichia coli (ETEC) adhesion to mammalian epithelial cells. Lactic acid bacteria (LAB) are known to be present abundantly in soybean tempeh. Some LAB species can produce exopolysaccharides (EPS) with anti-adhesion bioactivity against ETEC but there has been no report of anti-adhesion bioactive EPS from tempeh-associated LAB. We isolated EPS-producing LAB from tempeh-related sources, identified them, unambiguously elucidated their EPS structure and assessed the bioactivity of their EPS against ETEC. Pediococcus pentosaceus TL, Leuconostoc mesenteroides WA and L. mesenteroides WN produced both dextran (α-1,6 linked glucan; >1000 kDa) and levan (β-2,6 linked fructan; 650-760 kDa) in varying amounts and Leuconostoc citreum TR produced gel-forming α-1,6-mixed linkage dextran (829 kDa). All four isolates produced EPS that could adhere to ETEC cells and inhibit auto-aggregation of ETEC. EPS-PpTL, EPS-LmWA and EPS-LmWN were more bioactive towards pig-associated ETEC K88 while EPS-LcTR was more bioactive against human-associated ETEC H10407. Our finding is the first to report on the bioactivity of dextran against ETEC. Tempeh is a promising source of LAB isolates that can produce bioactive EPS against ETEC adhesion and aggregation.
Collapse
Affiliation(s)
- Theodorus Eko Pramudito
- Laboratory of Food Chemistry, Wageningen University & Research, P.O. Box 17, 6700 AA Wageningen, the Netherlands; Faculty of Biotechnology, Atma Jaya Catholic University of Indonesia, Indonesia
| | - Krishna Desai
- Laboratory of Food Chemistry, Wageningen University & Research, P.O. Box 17, 6700 AA Wageningen, the Netherlands; Marie Curie Early Stage Researcher, NutriLeads B.V., the Netherlands
| | - Camiel Voigt
- Food Microbiology, Wageningen University & Research, the Netherlands
| | - Eddy J Smid
- Food Microbiology, Wageningen University & Research, the Netherlands
| | - Henk A Schols
- Laboratory of Food Chemistry, Wageningen University & Research, P.O. Box 17, 6700 AA Wageningen, the Netherlands.
| |
Collapse
|
7
|
Zacharis C, Bonos E, Voidarou C(C, Magklaras G, Fotou K, Giannenas I, Giavasis I, Mitsagga C, Athanassiou C, Antonopoulou E, Grigoriadou K, Tzora A, Skoufos I. Combined Dietary Supplementation of Tenebrio molitor Larvae and Chitosan in Growing Pigs: A Pilot Study. Vet Sci 2024; 11:73. [PMID: 38393091 PMCID: PMC10893168 DOI: 10.3390/vetsci11020073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 01/14/2024] [Accepted: 02/01/2024] [Indexed: 02/25/2024] Open
Abstract
Nowadays, the global animal industry faces considerable challenges in securing sufficient feed resources. Responding to consumer demands for reduced use of antibiotics in animal nutrition, better animal welfare status, and reduced impact on the environment, there is an increased urgency to develop innovative functional feeds with a reduced environmental footprint and the ability to improve meat quality and safety. In an effort to explore innovative feed ingredients for growing pig diets, the combined dietary supplementation of Tenebrio molitor larvae and chitosan was investigated. An experimental trial was performed with 48 weaned pigs (34 days of life; mixed sex) that were randomly assigned to four treatment groups (with six males and six females each): Group A (control), Group B (supplemented with T. molitor larvae at 10%), Group C (supplemented with chitosan at 0.05%), and Group D (supplemented with both ingredients at 10% and 0.05%, respectively). On the 42nd day of the experimental trial, samples of blood, feces, and carcass parts were taken for analysis. The results indicated that the insect larvae meal significantly improved (p < 0.05) overall performance, increased (p < 0.05) blood red blood cell content, increased meat phenolic content (p < 0.05), improved meat oxidative stability (p < 0.05), and affected meat fatty acid profile (p < 0.05). On the other hand, chitosan had no significant effect on overall performance (p > 0.05), but it significantly increased blood lymphocyte content (p < 0.05), affected the fecal microbiota (p < 0.05), improved meat oxidative stability (p < 0.05), increased meat phenolic content (p < 0.05), and affected meat fatty acid composition (p < 0.05) and (p < 0.05) meat color. Finally, the combined use of both T. molitor and chitosan significantly affected some important zootechnical parameters (p < 0.05), fecal microbial populations (p < 0.05), meat color (p < 0.05), and fatty acid profile (p < 0.05). Further investigation into the potential interaction between insect larvae meals and chitosan in pig diets is advised.
Collapse
Affiliation(s)
- Christos Zacharis
- Department of Agriculture, University of Ioannina, Kostakioi Artas, 47100 Arta, Greece; (C.Z.); (E.B.); (C.V.); (G.M.); (K.F.); (A.T.); (I.S.)
| | - Eleftherios Bonos
- Department of Agriculture, University of Ioannina, Kostakioi Artas, 47100 Arta, Greece; (C.Z.); (E.B.); (C.V.); (G.M.); (K.F.); (A.T.); (I.S.)
| | - Chrysoula (Chrysa) Voidarou
- Department of Agriculture, University of Ioannina, Kostakioi Artas, 47100 Arta, Greece; (C.Z.); (E.B.); (C.V.); (G.M.); (K.F.); (A.T.); (I.S.)
| | - Georgios Magklaras
- Department of Agriculture, University of Ioannina, Kostakioi Artas, 47100 Arta, Greece; (C.Z.); (E.B.); (C.V.); (G.M.); (K.F.); (A.T.); (I.S.)
| | - Konstantina Fotou
- Department of Agriculture, University of Ioannina, Kostakioi Artas, 47100 Arta, Greece; (C.Z.); (E.B.); (C.V.); (G.M.); (K.F.); (A.T.); (I.S.)
| | - Ilias Giannenas
- School of Veterinary Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Ioannis Giavasis
- Department of Food Science and Nutrition, School of Agricultural Sciences, University of Thessaly, 43100 Karditsa, Greece; (I.G.); (C.M.)
| | - Chrysanthi Mitsagga
- Department of Food Science and Nutrition, School of Agricultural Sciences, University of Thessaly, 43100 Karditsa, Greece; (I.G.); (C.M.)
| | - Christos Athanassiou
- Department of Agriculture, Plant Production and Rural Environment, University of Thessaly, 38446 Nea Ionia, Greece;
| | - Efthimia Antonopoulou
- Department of Zoology, School of Biology, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
| | - Katerina Grigoriadou
- Institute of Plant Breeding and Genetic Resources, Hellenic Agricultural Organization (ELGO)-DIMITRA, 57001 Thessaloniki, Greece;
| | - Athina Tzora
- Department of Agriculture, University of Ioannina, Kostakioi Artas, 47100 Arta, Greece; (C.Z.); (E.B.); (C.V.); (G.M.); (K.F.); (A.T.); (I.S.)
| | - Ioannis Skoufos
- Department of Agriculture, University of Ioannina, Kostakioi Artas, 47100 Arta, Greece; (C.Z.); (E.B.); (C.V.); (G.M.); (K.F.); (A.T.); (I.S.)
| |
Collapse
|
8
|
Palkovicsné Pézsa N, Kovács D, Somogyi F, Karancsi Z, Móritz AV, Jerzsele Á, Rácz B, Farkas O. Effects of Lactobacillus rhamnosus DSM7133 on Intestinal Porcine Epithelial Cells. Animals (Basel) 2023; 13:3007. [PMID: 37835613 PMCID: PMC10571805 DOI: 10.3390/ani13193007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/27/2023] [Accepted: 09/22/2023] [Indexed: 10/15/2023] Open
Abstract
Antimicrobial resistance is one of the biggest health challenges nowadays. Probiotics are promising candidates as feed additives contributing to the health of the gastrointestinal tract. The beneficial effect of probiotics is species/strain specific; the potential benefits need to be individually assessed for each probiotic strain or species. We established a co-culture model, in which gastrointestinal infection was modeled using Escherichia coli (E. coli) and Salmonella enterica serovar Typhimurium (S. enterica serovar Typhimurium). Using intestinal porcine epithelial cells (IPEC-J2), the effects of pre-, co-, and post-treatment with Lactobacillus (L.) rhamnosus on the barrier function, intracellular (IC) reactive oxygen species (ROS) production, proinflammatory cytokine (IL-6 and IL-8) response, and adhesion inhibition were tested. E. coli- and S. Typhimurium-induced barrier impairment and increased ROS production could be counteracted using L. rhamnosus (p < 0.01). S. Typhimurium-induced IL-6 production was reduced via pre-treatment (p < 0.05) and post-treatment (p < 0.01); increased IL-8 secretion was decreased via pre-, co-, and post-treatment (p < 0.01) with L. rhamnosus. L. rhamnosus demonstrated significant inhibition of adhesion for both S. Typhimurium (p < 0.001) and E. coli (p < 0.001 in both pre-treatment and post-treatment; p < 0.05 in co-treatment). This study makes a substantial contribution to the understanding of the specific benefits of L. rhamnosus. Our findings can serve as a basis for further in vivo studies carried out in pigs and humans.
Collapse
Affiliation(s)
- Nikolett Palkovicsné Pézsa
- Department of Pharmacology and Toxicology, University of Veterinary Medicine Budapest, 1078 Budapest, Hungary; (D.K.); (F.S.); (Z.K.); (A.V.M.); (Á.J.); (O.F.)
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine Budapest, 1078 Budapest, Hungary
| | - Dóra Kovács
- Department of Pharmacology and Toxicology, University of Veterinary Medicine Budapest, 1078 Budapest, Hungary; (D.K.); (F.S.); (Z.K.); (A.V.M.); (Á.J.); (O.F.)
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine Budapest, 1078 Budapest, Hungary
| | - Fanni Somogyi
- Department of Pharmacology and Toxicology, University of Veterinary Medicine Budapest, 1078 Budapest, Hungary; (D.K.); (F.S.); (Z.K.); (A.V.M.); (Á.J.); (O.F.)
| | - Zita Karancsi
- Department of Pharmacology and Toxicology, University of Veterinary Medicine Budapest, 1078 Budapest, Hungary; (D.K.); (F.S.); (Z.K.); (A.V.M.); (Á.J.); (O.F.)
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine Budapest, 1078 Budapest, Hungary
| | - Alma Virág Móritz
- Department of Pharmacology and Toxicology, University of Veterinary Medicine Budapest, 1078 Budapest, Hungary; (D.K.); (F.S.); (Z.K.); (A.V.M.); (Á.J.); (O.F.)
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine Budapest, 1078 Budapest, Hungary
| | - Ákos Jerzsele
- Department of Pharmacology and Toxicology, University of Veterinary Medicine Budapest, 1078 Budapest, Hungary; (D.K.); (F.S.); (Z.K.); (A.V.M.); (Á.J.); (O.F.)
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine Budapest, 1078 Budapest, Hungary
| | - Bence Rácz
- Department of Anatomy and Histology, University of Veterinary Medicine Budapest, 1078 Budapest, Hungary;
| | - Orsolya Farkas
- Department of Pharmacology and Toxicology, University of Veterinary Medicine Budapest, 1078 Budapest, Hungary; (D.K.); (F.S.); (Z.K.); (A.V.M.); (Á.J.); (O.F.)
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine Budapest, 1078 Budapest, Hungary
| |
Collapse
|
9
|
Warner AJ, Tokach MD, Carrender B, Amachawadi RG, Labbé A, Heuser W, Coble K, DeRouchey JM, Woodworth JC, Goodband RD, Kalam R, Shi X, Nagaraja TG, Gebhardt JT. Evaluation of a Lactococcus lactis-based dried fermentation product administered through drinking water on nursery pig growth performance, fecal Escherichia coli virulence genes and pathotypes, antibiotic usage, and mortality. Transl Anim Sci 2023; 7:txad093. [PMID: 37649650 PMCID: PMC10465268 DOI: 10.1093/tas/txad093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Accepted: 07/29/2023] [Indexed: 09/01/2023] Open
Abstract
A total of 34,749 pigs were used in two experiments to evaluate the effects of a postbiotic dried fermentation product (DFP) administered through drinking water on nursery pig growth performance, antibiotic injection frequency, morbidity, mortality, fecal consistency, and characterization of fecal Escherichia coli. The DFP is composed of bioactive molecules derived from Lactococcus lactis. In Exp. 1, 350 barrows (DNA Line 200 × 400; initial body weight [BW] 6.1 ± 0.01 kg) were used in a 42-d study with five pigs per pen and 35 pens per treatment. The DFP was supplied for 14 d at a target dosage of 24 mg/kg BW using a water medicator at a 1:128 dilution. On days 7 and 14, fecal samples were collected for dry matter (DM) and to determine, by a multiplex polymerase chain reaction (PCR) assay, prevalence of 11 virulence genes characteristic of E. coli pathotypes. There was no evidence (P > 0.10) for differences for growth, incidence of diarrhea, number of antibiotic injections, removals, or fecal DM. On both fecal collection days, E. coli virulence genes were present with day 7 samples positive for genes that encode for hemolysins (hlyA, exhA), intimin (eae), and enteroaggregative heat-stable enterotoxin (astA). Prevalence of enterotoxin genes (elt, estA, estB, astA) increased on day 14, but DFP had no effects on the prevalence of any of the virulence genes. A total of 32 out of 72 E. coli isolates were identified as enterotoxigenic pathotype and all except one were from day 14 fecal samples. Fourteen isolates were positive for F4 fimbria and one isolate was positive for F4 and F18 fimbriae. In Exp. 2, 34,399 nursery pigs (initially 5.6 kg) were used in 20 nursery barns with 10 barns per treatment (control or DFP). The target dosage of the DFP for the first 14 d was 35 mg/kg BW. Following the 14-d supplementation period, pigs continued to be monitored for approximately 31 d. There was no evidence (P > 0.05) for the DFP to influence the overall percentage of pigs that died or growth performance. From days 0 to 14, providing the DFP reduced (P < 0.05) the percentage of pigs that were euthanized. However, providing the DFP increased (P < 0.05) the overall percentage of pigs that were euthanized and total mortality. For the number of antibiotic injections (treatment interventions), providing the DFP reduced the number of injections for the common period (P < 0.001) and overall (P = 0.002). These results indicate that the DFP did not influence growth performance but providing the DFP in Exp. 2 led to increased total nursery pig mortality.
Collapse
Affiliation(s)
- Alan J Warner
- Department of Animal Sciences and Industry, College of Agriculture, Kansas State University, Manhattan, KS 66506-0201, USA
| | - Mike D Tokach
- Department of Animal Sciences and Industry, College of Agriculture, Kansas State University, Manhattan, KS 66506-0201, USA
| | | | - Raghavendra G Amachawadi
- Department of Clinical Sciences, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506-0201, USA
| | | | | | - Kyle Coble
- JBS Live Pork, LLC, Greeley, CO 65101, USA
| | - Joel M DeRouchey
- Department of Animal Sciences and Industry, College of Agriculture, Kansas State University, Manhattan, KS 66506-0201, USA
| | - Jason C Woodworth
- Department of Animal Sciences and Industry, College of Agriculture, Kansas State University, Manhattan, KS 66506-0201, USA
| | - Robert D Goodband
- Department of Animal Sciences and Industry, College of Agriculture, Kansas State University, Manhattan, KS 66506-0201, USA
| | - Ramya Kalam
- Department of Clinical Sciences, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506-0201, USA
| | - Xiaorong Shi
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506-0201, USA
| | - T G Nagaraja
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506-0201, USA
| | - Jordan T Gebhardt
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506-0201, USA
| |
Collapse
|
10
|
Yuan B, Liu M, Luo S, Qu Q, Zhu M, Wang Z, Zhang X, Xie G, Li B, Wang W. ETEC regulates GPR109A expression in intestinal epithelial cells mediated by inflammatory factors secreted by macrophages. Res Vet Sci 2023; 154:15-21. [PMID: 36403332 DOI: 10.1016/j.rvsc.2022.11.005] [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: 07/21/2022] [Revised: 11/08/2022] [Accepted: 11/11/2022] [Indexed: 11/16/2022]
Abstract
Gut microbes control host immunity and homeostasis, and their abnormal changes are associated with the occurrence and development of diseases. GPR109A is an essential receptor on intestinal epithelial cells and interacts with gut microbes. Moreover, increased Enterotoxigenic Escherichia coli K88 strain colonization promotes GPR109A expression in vivo. This study evaluated the detailed mechanism of pathogenic bacteria promoting GPR109A expression. The results revealed that ETEC K88 indirectly fosters GPR109A expression in intestinal epithelial cells by stimulating the production of IL-1β and TNF-α through macrophages which are mediated by ERK1/2 pathway. The study explains the molecular mechanisms by which the bacteria regulate the homeostasis of the host intestinal gene expression during ETEC infection.
Collapse
Affiliation(s)
- Boyu Yuan
- Innovative Institute of Animal Healthy Breeding, College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China; State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Mingming Liu
- Innovative Institute of Animal Healthy Breeding, College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China
| | - Siyuan Luo
- Innovative Institute of Animal Healthy Breeding, College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China
| | - Qing Qu
- Innovative Institute of Animal Healthy Breeding, College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China
| | - Mingqiang Zhu
- Innovative Institute of Animal Healthy Breeding, College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China
| | - Zifan Wang
- Innovative Institute of Animal Healthy Breeding, College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China
| | - Xue Zhang
- Innovative Institute of Animal Healthy Breeding, College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China
| | - Gaijie Xie
- Innovative Institute of Animal Healthy Breeding, College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China
| | - Bai Li
- First clinical hospital of Jilin University, Changchun 130021, China.
| | - Wei Wang
- Innovative Institute of Animal Healthy Breeding, College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China.
| |
Collapse
|
11
|
Heat-labile enterotoxin enhances F4-producing enterotoxigenic E. coli adhesion to porcine intestinal epithelial cells by upregulating bacterial adhesins and STb enterotoxin. Vet Res 2022; 53:88. [PMID: 36303242 PMCID: PMC9615205 DOI: 10.1186/s13567-022-01110-4] [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: 07/07/2022] [Accepted: 09/19/2022] [Indexed: 11/10/2022] Open
Abstract
As one of the crucial enterotoxins secreted by enterotoxigenic Escherichia coli (ETEC), heat-labile enterotoxin (LT) enhances bacterial adherence both in vivo and in vitro; however, the underlying mechanism remains unclear. To address this, we evaluated the adherence of LT-producing and LT-deficient ETEC strains using the IPEC-J2 cell model. The expression levels of inflammatory cytokines and chemokines, and tight-junction proteins were evaluated in IPEC-J2 cells after infection with various ETEC strains. Further, the levels of adhesins and enterotoxins were also evaluated in F4ac-producing ETEC (F4 + ETEC) strains after treatment with cyclic AMP (cAMP). The adherence of the ΔeltAB mutant was decreased compared with the wild-type strain, whereas adherence of the 1836-2/pBR322-eltAB strain was markedly increased compared with the 1836-2 parental strain. Production of LT up-regulated the expression of TNF-α, IL-6, CXCL-8, and IL-10 genes. However, it did not appear to affect tight junction protein expression. Importantly, we found that cAMP leads to the upregulation of adhesin production and STb enterotoxin. Moreover, the F4 + ETEC strains treated with cAMP also had greater adhesion to IPEC-J2 cells, and the adherence of ΔfaeG, ΔfliC, and ΔestB mutants was decreased. These results indicate that LT enhances the adherence of F4 + ETEC due primarily to the upregulation of F4 fimbriae, flagellin, and STb enterotoxin expression and provide insights into the pathogenic mechanism of LT and ETEC.
Collapse
|
12
|
Xiao K, Yang Y, Zhang Y, Lv Q, Huang F, Wang D, Zhao J, Liu Y. Long-chain PUFA ameliorate enterotoxigenic Escherichia coli-induced intestinal inflammation and cell injury by modulating pyroptosis and necroptosis signaling pathways in porcine intestinal epithelial cells. Br J Nutr 2022; 128:835-850. [PMID: 34915950 DOI: 10.1017/s0007114521005092] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
This study was aimed to investigate whether EPA and arachidonic acid (ARA), the representative n-3 or n-6 PUFA, could alleviate enterotoxigenic Escherichia coli (ETEC) K88-induced inflammation and injury of intestinal porcine epithelial cells 1 (IPEC-1) by modulating pyroptosis and necroptosis signalling pathways. IPEC-1 cells were cultured with or without EPA or ARA in the presence or absence of ETEC K88. EPA and ARA reduced ETEC K88 adhesion and endotoxin content in the supernatant. EPA and ARA increased transepithelial electrical resistance, decreased permeability of fluorescein isothiocyanate-labelled dextran, increased membrane protein expression of occludin, ZO-1 and claudin-1 and relieved disturbed distribution of these proteins. EPA and ARA also reduced cell necrosis ratio. EPA or ARA reduced mRNA and concentration of TNF-α, IL-6 and IL-8 and decreased mRNA abundances of intestinal toll-like receptors 4 and its downstream signals. Moreover, EPA and ARA downregulated mRNA expression of nod-like receptor protein 3 (NLRP3), caspase 1 and IL-18 and inhibited protein expression of NLRP3, apoptosis-associated speck-like protein containing a CARD (ASC), gasdermin D and caspase-1. Finally, EPA and ARA reduced mRNA expression of fas-associated death domain protein, caspase 8, receptor-interacting protein kinase (RIP) 1, mixed lineage kinase-like protein (MLKL), phosphoglycerate mutase 5 (PGAM5), motility-related protein 1 (Drp1) and high mobility protein 1 (HMGB1) and inhibited protein expression of phosphorylated-RIP1, p-RIP3, p-MLKL and HMGB1. These data demonstrate that EPA and ARA prevent ETEC K88-induced cell inflammation and injury, which is partly through inhibiting pyroptosis and necroptosis signalling pathways.
Collapse
Affiliation(s)
- Kan Xiao
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan Polytechnic University, Wuhan, 430023, People's Republic of China
| | - Yang Yang
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan Polytechnic University, Wuhan, 430023, People's Republic of China
| | - Yang Zhang
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan Polytechnic University, Wuhan, 430023, People's Republic of China
| | - Qingqing Lv
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan Polytechnic University, Wuhan, 430023, People's Republic of China
| | - Feifei Huang
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan Polytechnic University, Wuhan, 430023, People's Republic of China
| | - Dan Wang
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan Polytechnic University, Wuhan, 430023, People's Republic of China
| | - Jiangchao Zhao
- Department of Animal Science, Division of Agriculture, University of Arkansas, Fayetteville, AR, USA
| | - Yulan Liu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan Polytechnic University, Wuhan, 430023, People's Republic of China
| |
Collapse
|
13
|
Lactoferrin Decreases Enterotoxigenic Escherichia coli-Induced Fluid Secretion and Bacterial Adhesion in the Porcine Small Intestine. Pharmaceutics 2022; 14:pharmaceutics14091778. [PMID: 36145526 PMCID: PMC9504966 DOI: 10.3390/pharmaceutics14091778] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/22/2022] [Accepted: 08/23/2022] [Indexed: 11/25/2022] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) infections are one of the most prevalent causes of post-weaning diarrhea in piglets, resulting in morbidity, mortality and elevated use of antibiotics. The emergence and further spread of antimicrobial resistance together with the growing demand for high quality animal protein requires the identification of novel alternatives for antimicrobials. A promising alternative is lactoferrin, as we previously showed that it can both inhibit the growth and degrade bacterial virulence factors of porcine ETEC strains in vitro. Aiming to confirm these findings in vivo, we performed a small intestinal segment perfusion experiment in piglets. Here, we showed that lactoferrin could not only decrease ETEC-induced fluid secretion, but also their ability to colonize the small intestinal epithelium. Furthermore, while ETEC infection induced pro-inflammatory cytokine mRNA expression in this experiment, lactoferrin was not able to counteract these responses. In addition, a bacterial motility assay showed that lactoferrin can reduce the motility of ETEC. Our findings further support the use of lactoferrin as an alternative for antimicrobials and also show its potential for the prevention of ETEC infections in pigs.
Collapse
|
14
|
The Impact of Quercetin and Its Methylated Derivatives 3-o-Methylquercetin and Rhamnazin in Lipopolysaccharide-Induced Inflammation in Porcine Intestinal Cells. Antioxidants (Basel) 2022; 11:antiox11071265. [PMID: 35883756 PMCID: PMC9312192 DOI: 10.3390/antiox11071265] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/18/2022] [Accepted: 06/23/2022] [Indexed: 12/04/2022] Open
Abstract
Oxidative stress in the small intestine can lead to inflammation and barrier malfunction. The present study describes the effect of quercetin (Q), 3-o-methylquercetin (QM), and rhamnazin (R) on cell viability, paracellular permeability, production of intracellular reactive oxygen species (ROS), extracellular hydrogen peroxide (H2O2), and interleukin-6 (IL-6) after challenging jejunal cells (IPEC-J2) with different types (Salmonella enterica ser. Typhimurium, Escherichia coli O111:B4, and E. coli O127:B8) of lipopolysaccharides (LPS) applied in 10 µg/mL concentration. The intracellular ROS level increased after all LPS treatments, which could be decreased by all tested flavonoid compounds in 50 µM concentration. Extracellular H2O2 production significantly increased after Q and R treatment (50 µM). S. Typhimurium LPS could significantly increase IL-6 production of enterocytes, which could be alleviated by Q, QM, and R (50 µM) as well. Using fluorescein isothiocyanate dextran (FD4) tracer dye, we could demonstrate that S. Typhimurium LPS significantly increased the permeability of the cell layer. The simultaneous treatments of S. Typhimurium LPS and the flavonoid compounds showed no alteration in FD4 penetration compared to untreated cells. These results highlight that Q, QM, and R are promising substances that can be used to protect intestinal epithelial cells from the deteriorating effects of oxidative stress.
Collapse
|
15
|
Intestinal Epithelial Cells Modulate the Production of Enterotoxins by Porcine Enterotoxigenic E. coli Strains. Int J Mol Sci 2022; 23:ijms23126589. [PMID: 35743033 PMCID: PMC9223395 DOI: 10.3390/ijms23126589] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 06/03/2022] [Accepted: 06/12/2022] [Indexed: 01/23/2023] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) strains are one of the most common etiological agents of diarrhea in both human and farm animals. In addition to encoding toxins that cause diarrhea, ETEC have evolved numerous strategies to interfere with host defenses. These strategies most likely depend on the sensing of host factors, such as molecules secreted by gut epithelial cells. The present study tested whether the exposure of ETEC to factors secreted by polarized IPEC-J2 cells resulted in transcriptional changes of ETEC-derived virulence factors. Following the addition of host-derived epithelial factors, genes encoding enterotoxins, secretion-system-associated proteins, and the key regulatory molecule cyclic AMP (cAMP) receptor protein (CRP) were substantially modulated, suggesting that ETEC recognize and respond to factors produced by gut epithelial cells. To determine whether these factors were heat sensitive, the IEC-conditioned medium was incubated at 56 °C for 30 min. In most ETEC strains, heat treatment of the IEC-conditioned medium resulted in a loss of transcriptional modulation. Taken together, these data suggest that secreted epithelial factors play a role in bacterial pathogenesis by modulating the transcription of genes encoding key ETEC virulence factors. Further research is warranted to identify these secreted epithelial factors and how ETEC sense these molecules to gain a competitive advantage in the early engagement of the gut epithelium.
Collapse
|
16
|
Significance of Mucosa-Associated Microbiota and Its Impacts on Intestinal Health of Pigs Challenged with F18 +E. coli. Pathogens 2022; 11:pathogens11050589. [PMID: 35631110 PMCID: PMC9145386 DOI: 10.3390/pathogens11050589] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 05/05/2022] [Accepted: 05/14/2022] [Indexed: 01/27/2023] Open
Abstract
The objective of this study was to evaluate the significance of jejunal mucosa-associated microbiota and its impacts on the intestinal health of pigs challenged with F18+ Escherichia coli. Forty-four newly-weaned pigs were allotted to two treatments in a randomized complete block design with sex as blocks. Pigs were fed common diets for 28 d. At d 7 post-weaning, pigs were orally inoculated with saline solution or F18+ E. coli. At d 21 post-challenge, feces and blood were collected and pigs were euthanized to collect jejunal tissue to evaluate microbiota and intestinal health parameters. The relative abundance of Firmicutes and Bacteroidetes was lower (p < 0.05) in jejunal mucosa than in feces, whereas Proteobacteria was greater (p < 0.05) in jejunal mucosa. F18+ E. coli increased (p < 0.05) protein carbonyl, Helicobacteraceae, Pseudomonadaceae, Xanthomonadaceae, and Peptostreptococcaceae and reduced (p < 0.05) villus height, Enterobacteriaceae, Campylobacteraceae, Brachyspiraceae, and Caulobacteraceae in jejunal mucosa, whereas it reduced (p < 0.05) Spirochaetaceae and Oscillospiraceae in feces. Collectively, jejunal mucosa-associated microbiota differed from those in feces. Compared with fecal microbiota, the change of mucosa-associated microbiota by F18+ E. coli was more prominent, and it was mainly correlated with increased protein carbonyl and reduced villus height in jejunal mucosa impairing the intestinal health of nursery pigs.
Collapse
|
17
|
Effects of Bacillus licheniformis and Bacillus subtilis on Gut Barrier Function, Proinflammatory Response, ROS Production and Pathogen Inhibition Properties in IPEC-J2—Escherichia coli/Salmonella Typhimurium Co-Culture. Microorganisms 2022; 10:microorganisms10050936. [PMID: 35630380 PMCID: PMC9145911 DOI: 10.3390/microorganisms10050936] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 04/23/2022] [Accepted: 04/27/2022] [Indexed: 02/04/2023] Open
Abstract
The emergence of antimicrobial resistance raises serious concerns worldwide. Probiotics offer a promising alternative to enhance growth promotion in farm animals; however, their mode of action still needs to be elucidated. The IPEC-J2 cell line (porcine intestinal epithelial cells) is an appropriate tool to study the effect of probiotics on intestinal epithelial cells. In our experiments, IPEC-J2 cells were challenged by two gastrointestinal (GI) infection causing agents, Escherichia coli (E. coli) or Salmonella enterica ser. Typhimurium (S. Typhimurium). We focused on determining the effect of pre-, co-, and post-treatment with two probiotic candidates, Bacillus licheniformis or Bacillus subtilis, on the barrier function, proinflammatory cytokine (IL-6 and IL-8) response, and intracellular reactive oxygen species (ROS) production of IPEC-J2 cells, in addition to the adhesion inhibition effect. Bacillus licheniformis (B. licheniformis) and Bacillus subtilis (B. subtilis) proved to be anti-inflammatory and had an antioxidant effect under certain treatment combinations, and further effectively inhibited the adhesion of pathogenic bacteria. Interestingly, they had little effect on paracellular permeability. Based on our results, Bacillus licheniformis and Bacillus subtilis are both promising candidates to contribute to the beneficial effects of probiotic multispecies mixtures.
Collapse
|
18
|
Palkovicsné Pézsa N, Kovács D, Gálfi P, Rácz B, Farkas O. Effect of Enterococcus faecium NCIMB 10415 on Gut Barrier Function, Internal Redox State, Proinflammatory Response and Pathogen Inhibition Properties in Porcine Intestinal Epithelial Cells. Nutrients 2022; 14:nu14071486. [PMID: 35406099 PMCID: PMC9002907 DOI: 10.3390/nu14071486] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/30/2022] [Accepted: 04/01/2022] [Indexed: 01/14/2023] Open
Abstract
In farm animals, intestinal diseases caused by Salmonella spp. and Escherichia coli may lead to significant economic loss. In the past few decades, the swine industry has largely relied on the prophylactic use of antibiotics to control gastrointestinal diseases. The development of antibiotic resistance has become an important issue both in animal and human health. The use of antibiotics for prophylactic purposes has been banned, moreover the new EU regulations further restrict the application of antibiotics in veterinary use. The swine industry seeks alternatives that are capable of maintaining the health of the gastrointestinal tract. Probiotics offer a promising alternative; however, their mode of action is not fully understood. In our experiments, porcine intestinal epithelial cells (IPEC-J2 cells) were challenged by Salmonella Typhimurium or Escherichia coli and we aimed at determining the effect of pre-, co-, and post-treatment with Enterococcus faecium NCIMB 10415 on the internal redox state, paracellular permeability, IL-6 and IL-8 secretion of IPEC-J2 cells. Moreover, the adhesion inhibition effect was also investigated. Enterococcus faecium was able to reduce oxidative stress and paracellular permeability of IPEC-J2 cells and could inhibit the adhesion of Salmonella Typhimurium and Escherichia coli. Based on our results, Enterococcus faecium is a promising candidate to maintain the health of the gastrointestinal tract.
Collapse
Affiliation(s)
- Nikolett Palkovicsné Pézsa
- Department of Pharmacology and Toxicology, University of Veterinary Medicine Budapest, 1078 Budapest, Hungary; (D.K.); (P.G.); (O.F.)
- Correspondence:
| | - Dóra Kovács
- Department of Pharmacology and Toxicology, University of Veterinary Medicine Budapest, 1078 Budapest, Hungary; (D.K.); (P.G.); (O.F.)
| | - Péter Gálfi
- Department of Pharmacology and Toxicology, University of Veterinary Medicine Budapest, 1078 Budapest, Hungary; (D.K.); (P.G.); (O.F.)
| | - Bence Rácz
- Department of Anatomy and Histology, University of Veterinary Medicine Budapest, 1078 Budapest, Hungary;
| | - Orsolya Farkas
- Department of Pharmacology and Toxicology, University of Veterinary Medicine Budapest, 1078 Budapest, Hungary; (D.K.); (P.G.); (O.F.)
| |
Collapse
|
19
|
Ge L, Liu D, Mao X, Liu S, Guo J, Hou L, Chen X, Huang K. Low Dose of Deoxynivalenol Aggravates Intestinal Inflammation and Barrier Dysfunction Induced by Enterotoxigenic Escherichia coli Infection through Activating Macroautophagy/NLRP3 Inflammasomes. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:3009-3022. [PMID: 35201764 DOI: 10.1021/acs.jafc.1c07834] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The toxicity of deoxynivalenol (DON) in healthy humans and animals has been extensively studied. However, whether the natural-low-dose DON is scatheless under unhealthy conditions, especially intestinal injury, is unknown. Infection of enterotoxigenic Escherichia coli (ETEC) is a classical intestinal injury model. In this study, we explored the effects of low-dose DON on intestinal injury induced by the ETEC infection and the underlying mechanism in piglets, mice, and IPEC-J2 monolayer cells. Results showed that significant growth slowdown, severe diarrhea, and intestinal damage, bacterial multiplication, and translocation were observed in the experimental group (low-dose DON, 0.75 mg/kg in feed for piglets, and 1 mg/kg body weight for mice, combined with the ETEC infection). Meanwhile, more aggressive intestinal inflammation and barrier dysfunction were observed in animals and IPEC-J2 monolayer cells. Higher expression levels of NLRP3 inflammasome and LC3B were observed in jejunum and IPEC-J2 in the experimental group. After treatment with NLRP3 or caspase1 inhibitors, excessive intestinal inflammation rather than barrier dysfunction in the experimental group was limited. CRISPR-Cas9-mediated knockout of LC3B alleviated intestinal inflammation and barrier dysfunction and also inhibited NLRP3 inflammasome. In conclusion, a low dose of DON aggravates intestinal inflammation and barrier dysfunction induced by the ETEC infection by activating macroautophagy and NLRP3 inflammasome.
Collapse
Affiliation(s)
- 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
| | - Dandan 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
| | - 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
| | - 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
| | - Junyan Guo
- 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
| | - 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
| | - 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
|
20
|
Fiil BK, Thrane SW, Pichler M, Kittilä T, Ledsgaard L, Ahmadi S, Maigaard Hermansen GM, Jelsbak L, Lauridsen C, Brix S, Laustsen AH. Orally-active bivalent VHH construct prevents proliferation of F4+ enterotoxigenic Escherichia coli in weaned piglets. iScience 2022; 25:104003. [PMID: 35310945 PMCID: PMC8931358 DOI: 10.1016/j.isci.2022.104003] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 12/21/2021] [Accepted: 02/25/2022] [Indexed: 11/25/2022] Open
Abstract
A major challenge in industrial pig production is the prevalence of post-weaning diarrhea (PWD) in piglets, often caused by enterotoxigenic Escherichia coli (ETEC). The increased use of antibiotics and zinc oxide to treat PWD has raised global concerns regarding antimicrobial resistance development and environmental pollution. Still, alternative treatments targeting ETEC and counteracting PWD are largely lacking. Here, we report the design of a pH, temperature, and protease-stable bivalent VHH-based protein BL1.2 that cross-links a F4+ ETEC model strain by selectively binding to its fimbriae. This protein inhibits F4+ ETEC adhesion to porcine epithelial cells ex vivo and decreases F4+ ETEC proliferation when administrated as a feed additive to weaned F4+ ETEC challenged piglets. These findings highlight the potential of a highly specific bivalent VHH-based feed additive in effectively delimiting pathogenic F4+ ETEC bacteria proliferation in piglets and may represent a sustainable solution for managing PWD while circumventing antimicrobial resistance development. A binding protein was designed as a bivalent VHH construct with a (GGGGS)3 linker The protein can cross-link F4+ enterotoxigenic Escherichia coli (ETEC) in vitro The protein can prevent adhesion of F4+ ETEC to porcine epithelial cells ex vivo The protein can prevent proliferation of F4+ ETEC in piglets
Collapse
|
21
|
Kim K, He Y, Jinno C, Kovanda L, Li X, Bravo D, Cox E, Liu Y. Supplementation of oligosaccharide-based polymer enhanced growth and disease resistance of weaned pigs by modulating intestinal integrity and systemic immunity. J Anim Sci Biotechnol 2022; 13:10. [PMID: 35016715 PMCID: PMC8753815 DOI: 10.1186/s40104-021-00655-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 11/21/2021] [Indexed: 11/18/2022] Open
Abstract
Background There is a great demand for antibiotic alternatives to maintain animal health and productivity. The objective of this experiment was to determine the efficacy of dietary supplementation of a blood group A6 type 1 antigen oligosaccharides-based polymer (Coligo) on growth performance, diarrhea severity, intestinal health, and systemic immunity of weaned pigs experimentally infected with an enterotoxigenic Escherichia coli (ETEC), when compared with antibiotics. Results Pigs in antibiotic carbadox or Coligo treatment groups had greater (P < 0.05) body weight on d 5 or d 11 post-inoculation (PI) than pigs in the control group, respectively. Supplementation of antibiotics or Coligo enhanced (P < 0.05) feed efficiency from d 0 to 5 PI and reduced (P < 0.05) frequency of diarrhea throughout the experiment, compared with pigs in the control group. Supplementation of antibiotics reduced (P < 0.05) fecal β-hemolytic coliforms on d 2, 5, and 8 PI. Pigs in antibiotics or Coligo groups had reduced (P < 0.05) neutrophil counts and serum haptoglobin concentration compared to pigs in the control group on d 2 and 5 PI. Pigs in Coligo had reduced (P < 0.05) total coliforms in mesenteric lymph nodes on d 5 and 11 PI, whereas pigs in antibiotics or Coligo groups had reduced (P < 0.05) total coliforms in spleen on d 11 PI compared with pigs in the control group. On d 5 PI, pigs in the Coligo group had greater (P < 0.05) gene expression of ZO1 in jejunal mucosa, but less (P < 0.05) mRNA expression of IL1B, IL6, and TNF in ileal mucosa, in comparison with pigs in the control group. Supplementation of antibiotics enhanced (P < 0.05) the gene expression of OCLN in jejunal mucosa but decreased (P < 0.05) IL1B and IL6 gene expression in ileal mucosa, compared with the control. On d 11 PI, supplementation of antibiotics or Coligo up-regulated (P < 0.05) gene expression of CLDN1 in jejunal mucosa, but Coligo reduced (P < 0.05) IL6 gene expression in ileal mucosa compared to pigs in the control group. Conclusions Supplementation of Coligo improved growth performance, alleviated diarrhea severity, and enhanced gut health in weaned pigs infected with ETEC F18 in a manner similar to in-feed antibiotics. Supplementary Information The online version contains supplementary material available at 10.1186/s40104-021-00655-2.
Collapse
Affiliation(s)
- Kwangwook Kim
- Department of Animal Science, University of California, Davis, CA, 95616, USA
| | - Yijie He
- Department of Animal Science, University of California, Davis, CA, 95616, USA
| | - Cynthia Jinno
- Department of Animal Science, University of California, Davis, CA, 95616, USA
| | - Lauren Kovanda
- Department of Animal Science, University of California, Davis, CA, 95616, USA
| | - Xunde Li
- School of Veterinary Medicine, University of California, Davis, CA, 95616, USA
| | | | - Eric Cox
- Department of Virology, Parasitology and Immunology, Ghent University, 9000, Ghent, Belgium
| | - Yanhong Liu
- Department of Animal Science, University of California, Davis, CA, 95616, USA.
| |
Collapse
|
22
|
Duarte ME, Kim SW. Intestinal microbiota and its interaction to intestinal health in nursery pigs. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2022; 8:169-184. [PMID: 34977387 PMCID: PMC8683651 DOI: 10.1016/j.aninu.2021.05.001] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 04/20/2021] [Accepted: 05/07/2021] [Indexed: 02/07/2023]
Abstract
The intestinal microbiota has gained increased attention from researchers within the swine industry due to its role in promoting intestinal maturation, immune system modulation, and consequently the enhancement of the health and growth performance of the host. This review aimed to provide updated scientific information on the interaction among intestinal microbiota, dietary components, and intestinal health of pigs. The small intestine is a key site to evaluate the interaction of the microbiota, diet, and host because it is the main site for digestion and absorption of nutrients and plays an important role within the immune system. The diet and its associated components such as feed additives are the main factors affecting the microbial composition and is central in stimulating a beneficial population of microbiota. The microbiota–host interaction modulates the immune system, and, concurrently, the immune system helps to modulate the microbiota composition. The direct interaction between the microbiota and the host is an indication that the mucosa-associated microbiota can be more effective in evaluating its effect on health parameters. It was demonstrated that the mucosa-associated microbiota should be evaluated when analyzing the interaction among diets, microbiota, and health. In addition, supplementation of feed additives aimed to promote the intestinal health of pigs should consider their roles in the modulation of mucosa-associated microbiota as biomarkers to predict the response of growth performance to dietary interventions.
Collapse
Affiliation(s)
- Marcos Elias Duarte
- Department of Animal Science, North Carolina State University, Raleigh, NC, 27695, United States
| | - Sung Woo Kim
- Department of Animal Science, North Carolina State University, Raleigh, NC, 27695, United States
| |
Collapse
|
23
|
Li H, Shang Z, Liu X, Qiao Y, Wang K, Qiao J. Clostridium butyricum Alleviates Enterotoxigenic Escherichia coli K88 -Induced Oxidative Damage Through Regulating the p62-Keap1-Nrf2 Signaling Pathway and Remodeling the Cecal Microbial Community. Front Immunol 2021; 12:771826. [PMID: 34899723 PMCID: PMC8660075 DOI: 10.3389/fimmu.2021.771826] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 10/28/2021] [Indexed: 12/21/2022] Open
Abstract
Clostridium butyricum (CB) can enhance antioxidant capacity and alleviate oxidative damage, but the molecular mechanism by which this occurs remains unclear. This study used enterotoxigenic Escherichia coli (ETEC) K88 as a pathogenic model, and the p62-Keap1-Nrf2 signaling pathway and intestinal microbiota as the starting point to explore the mechanism through which CB alleviates oxidative damage. After pretreatment with CB for 15 d, mice were challenged with ETEC K88 for 24 h. The results suggest that CB pretreatment can dramatically reduce crypt depth (CD) and significantly increase villus height (VH) and VH/CD in the jejunum of ETEC K88-infected mice and relieve morphological lesions of the liver and jejunum. Additionally, compared with ETEC-infected group, pretreatment with 4.4×106 CFU/mL CB can significantly reduce malondialdehyde (MDA) level and dramatically increase superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px) levels in the serum. This pretreatment can also greatly increase the mRNA expression levels of tight junction proteins and genes related to the p62-Keap1-Nrf2 signaling pathway in the liver and jejunum in ETEC K88-infected mice. Meanwhile, 16S rDNA amplicon sequencing revealed that Clostridium disporicum was significantly enriched after ETEC K88 challenge relative to the control group, while Lactobacillus was significantly enriched after 4.4×106 CFU/mL CB treatment. Furthermore, 4.4×106 CFU/mL CB pretreatment increased the short-chain fatty acid (SCFA) contents in the cecum of ETEC K88-infected mice. Moreover, we found that Lachnoclostridium, Roseburia, Lactobacillus, Terrisporobacter, Akkermansia, and Bacteroides are closely related to SCFA contents and oxidative indicators. Taken together, 4.4×106 CFU/mL CB pretreatment can alleviate ETEC K88-induced oxidative damage through activating the p62-Keap1-Nrf2 signaling pathway and remodeling the cecal microbiota community in mice.
Collapse
Affiliation(s)
- Haihua Li
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin, China
| | - Zhiyuan Shang
- Tianjin Key Laboratory of Animal and Plant Resistance, College of Life Sciences, Tianjin Normal University, Tianjin, China
| | - Xuejiao Liu
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin, China
| | - Yingying Qiao
- Faculty of Biology and Technology, Sumy National Agrarian University, Sumy, Ukraine
| | - Kewei Wang
- Tianjin Key Laboratory of Animal and Plant Resistance, College of Life Sciences, Tianjin Normal University, Tianjin, China
| | - Jiayun Qiao
- Tianjin Key Laboratory of Animal and Plant Resistance, College of Life Sciences, Tianjin Normal University, Tianjin, China
| |
Collapse
|
24
|
Saccharomyces Cerevisiae Var Boulardii CNCM I-1079 Reduces Expression of Genes Involved in Inflammatory Response in Porcine Cells Challenged by Enterotoxigenic E. Coli and Influences Bacterial Communities in an In Vitro Model of the Weaning Piglet Colon. Antibiotics (Basel) 2021; 10:antibiotics10091101. [PMID: 34572682 PMCID: PMC8467900 DOI: 10.3390/antibiotics10091101] [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: 08/13/2021] [Revised: 09/06/2021] [Accepted: 09/07/2021] [Indexed: 11/17/2022] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) is the main infectious agent responsible for piglet post-weaning diarrhea with high mortality rates. Antimicrobials represent the current principal strategy for treating ETEC infections in pig farms, but the occurrence of multi-resistant bacterial strains has considerably increased in the last decades. Thus, finding non-antibiotic alternatives becomes a real emergency. In this context, we investigated the effect of a live yeast strain, Saccharomyces cerevisiae var boulardii CNCM I-1079 (SB) in an in vitro model of the weaning piglet colon implemented with a mucus phase (MPigut-IVM) inoculated with ETEC and coupled with an intestinal porcine cell line IPI-2I. We showed that SB was able to modulate the in vitro microbiota through an increase in Bacteroidiaceae and a decrease in Prevotellaceae families. Effluents collected from the SB treated bioreactors were able to mitigate the expression level of genes encoding non-gel forming mucins, tight junction proteins, innate immune pathway, and pro-inflammatory response in IPI-2I cells. Furthermore, SB exerted a significant protective effect against ETEC adhesion on porcine IPEC-J2 intestinal cells in a dose-dependent manner and showed a positive effect on ETEC-challenged IPEC-J2 by lowering expression of genes involved in pro-inflammatory immune responses. Our results showed that the strain SB CNCM I-1079 could prevent microbiota dysbiosis associated with weaning and protect porcine enterocytes from ETEC infections by reducing bacterial adhesion and modulating the inflammatory response.
Collapse
|
25
|
Chitosan-chelated zinc modulates ileal microbiota, ileal microbial metabolites, and intestinal function in weaned piglets challenged with Escherichia coli K88. Appl Microbiol Biotechnol 2021; 105:7529-7544. [PMID: 34491402 DOI: 10.1007/s00253-021-11496-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 07/16/2021] [Accepted: 07/20/2021] [Indexed: 01/17/2023]
Abstract
This study was to investigate the effects of chitosan-chelated zinc on ileal microbiota, inflammatory response, and barrier function in weaned piglets challenged with Escherichia coli K88. Piglets of the chitosan-chelated zinc treatment (Cs-Zn; 100 mg zinc + 766 mg chitosan/kg basal diet, from chitosan-chelated zinc) and the chitosan treatment (CS, 766 mg chitosan/kg basal diet) had significantly increased ileal villus height and the ratio of villi height to crypt depth. CS-Zn group piglets had a higher abundance of Lactobacillus in the ileal digesta, while the abundance of Streptococcus, Escherichia shigella, Actinobacillus, and Clostridium sensu stricto 6 was significantly decreased. The concentrations of propionate, butyrate, and lactate in the CS-Zn group piglets were significantly increased, while the pH value was significantly decreased. Furthermore, the concentrations of IL-1β, TNF-α, MPO, and INF-γ in the ileal mucosa of the CS-Zn and the H-ZnO group (pharmacological dose of 1600 mg Zn/kg basal diet, from ZnO) were significantly lower than those of the control group fed with basal diet, and the mRNA expression of TLR4, MyD88, and NF-κB of the CS-Zn group was also reduced. In addition, the mRNA expression of IGF-1 was increased, the protein expression of occludin and claudin-1 was enhanced, while the mRNA expression of caspase 3 and caspase 8 was decreased in the CS-Zn group. These results suggest CS-Zn treatment could help modulate the composition of ileal microbiota, attenuate inflammatory response, and maintain the intestinal function in weaned piglets challenged with Escherichia coli K88. KEY POINTS: • Chitosan-chelated zinc significantly modulated ileal microbiota. • Chitosan-chelated zinc can improve ileal health. • The ileal microbiota plays an important role in host health.
Collapse
|
26
|
Different Responses of Microbiota across Intestinal Tract to Enterococcus faecium HDRsEf1 and Their Correlation with Inflammation in Weaned Piglets. Microorganisms 2021; 9:microorganisms9081767. [PMID: 34442847 PMCID: PMC8402050 DOI: 10.3390/microorganisms9081767] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 08/11/2021] [Accepted: 08/16/2021] [Indexed: 12/16/2022] Open
Abstract
Enterococcus faecium HDRsEf1 (HDRsEf1) was identified to reduce the incidence of diarrhea in weaned piglets, but the mechanism has not been elucidated yet. Based on the fact that gut microbiota plays a crucial role in regulating inflammatory responses, the effects of HDRsEf1 on microbiota across the intestinal tract in weaned piglets were investigated. Microbiota from the luminal contents and the mucosa of the ileum, cecum, and colon of HDRsEf1-treated piglets were explored by 16S rRNA sequencing and qPCR. It was demonstrated that microbiota in different gut niches responded specifically to HDRsEf1, with major alterations occurring in the ileum and cecum. The total bacterial load of microbiota in ileal luminal contents and the relative abundance of Escherichia-Shigella in the ileal mucosa was significantly down-regulated by HDRsEf1 administration, while the relative abundance of butyrate-producing bacteria (including Clostridiaceae-1, Rumencoccidae, and Erysipelotrichaceae) in cecal luminal contents was significantly up-regulated. Moreover, the utilization of HDRsEf1 improved intestinal morphological development and reduced the inflammatory response, which were negatively correlated with the relative abundance of Escherichia-Shigella in the ileal mucosa and butyrate-producing bacteria in cecal luminal contents, respectively. Collectively, this study suggests that the administration of HDRsEf1 alters gut microbiota, thereby alleviating inflammation and improving intestinal morphological development in weaned piglets.
Collapse
|
27
|
Vermeire B, Gonzalez LM, Jansens RJJ, Cox E, Devriendt B. Porcine small intestinal organoids as a model to explore ETEC-host interactions in the gut. Vet Res 2021; 52:94. [PMID: 34174960 PMCID: PMC8235647 DOI: 10.1186/s13567-021-00961-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 06/03/2021] [Indexed: 12/14/2022] Open
Abstract
Small intestinal organoids, or enteroids, represent a valuable model to study host–pathogen interactions at the intestinal epithelial surface. Much research has been done on murine and human enteroids, however only a handful studies evaluated the development of enteroids in other species. Porcine enteroid cultures have been described, but little is known about their functional responses to specific pathogens or their associated virulence factors. Here, we report that porcine enteroids respond in a similar manner as in vivo gut tissues to enterotoxins derived from enterotoxigenic Escherichia coli, an enteric pathogen causing postweaning diarrhoea in piglets. Upon enterotoxin stimulation, these enteroids not only display a dysregulated electrolyte and water balance as shown by their swelling, but also secrete inflammation markers. Porcine enteroids grown as a 2D-monolayer supported the adhesion of an F4+ ETEC strain. Hence, these enteroids closely mimic in vivo intestinal epithelial responses to gut pathogens and are a promising model to study host–pathogen interactions in the pig gut. Insights obtained with this model might accelerate the design of veterinary therapeutics aimed at improving gut health.
Collapse
Affiliation(s)
- Bjarne Vermeire
- Department of Virology, Parasitology, Immunology, Faculty of Veterinary Medicine, Laboratory of Immunology, Ghent University, 9820, Merelbeke, Belgium
| | - Liara M Gonzalez
- Laboratory of Intestinal Regenerative Medicine, College of Veterinary Medicine, NCSU, Raleigh, NC, USA
| | - Robert J J Jansens
- Department of Virology, Parasitology, Immunology, Faculty of Veterinary Medicine, Laboratory of Immunology, Ghent University, 9820, Merelbeke, Belgium
| | - Eric Cox
- Department of Virology, Parasitology, Immunology, Faculty of Veterinary Medicine, Laboratory of Immunology, Ghent University, 9820, Merelbeke, Belgium
| | - Bert Devriendt
- Department of Virology, Parasitology, Immunology, Faculty of Veterinary Medicine, Laboratory of Immunology, Ghent University, 9820, Merelbeke, Belgium.
| |
Collapse
|
28
|
Yang KM, Zhu C, Wang L, Cao ST, Yang XF, Gao KG, Jiang ZY. Early supplementation with Lactobacillus plantarum in liquid diet modulates intestinal innate immunity through toll-like receptor 4-mediated mitogen-activated protein kinase signaling pathways in young piglets challenged with Escherichia coli K88. J Anim Sci 2021; 99:6259343. [PMID: 33928383 DOI: 10.1093/jas/skab128] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 04/19/2021] [Indexed: 12/12/2022] Open
Abstract
This study was conducted to investigate the effects of early supplementation during 4 to 18 d of age with Lactobacillus plantarum (LP) in liquid diets on intestinal innate immune response in young piglets infected with enterotoxigenic Escherichia coli (ETEC) K88. Seventy-two barrow piglets at 4 d old were assigned to basal or LP-supplemented liquid diet (5 × 1010 CFU·kg-1). On day 15, piglets from each group were orally challenged with either ETEC K88 (1 × 108 CFU·kg-1) or the same amount of phosphate-buffered saline. The intestinal mucosa, mesenteric lymph node (MLN), and spleen samples were collected on day 18. Here, we found that LP pretreatment significantly decreased the mRNA relative expression of inflammatory cytokines (interleukin [IL]-1β, IL-8, and tumor necrosis factor-α), porcine β-defensin 2 (pBD-2), and mucins (MUC1 and MUC4) in the jejunal mucosa in piglets challenged with ETEC K88 (P < 0.05). Moreover, LP significantly decreased the ileal mucosa mRNA relative expression of IL-8 and MUC4 in young piglets challenged with ETEC K88 (P < 0.05). Furthermore, the piglets of the LP + ETEC K88 group had lower protein levels of IL-8, secretory immunoglobulin A, pBD-2, and MUC4 in the jejunal mucosa than those challenged with ETEC K88 (P < 0.05). Besides, LP supplementation reduced the percentage of gamma/delta T cells receptor (γδTCR) and CD172a+ (SWC3+) cells in MLN and the percentage of γδTCR cells in the spleen of young piglets after the ETEC K88 challenge. Supplementation with LP in liquid diets prevented the upregulated protein abundance of toll-like receptor (TLR) 4, phosphorylation-p38, and phosphorylation-extracellular signal-regulated protein kinases in the jejunal mucosa induced by ETEC K88 (P < 0.05). In conclusion, LP supplementation in liquid diet possesses anti-inflammatory activity and modulates the intestinal innate immunity during the early life of young piglets challenged with ETEC K88, which might be attributed to the suppression of TLR4-mediated mitogen-activated protein kinase signaling pathways. Early supplementation with LP in liquid diets regulates the innate immune response, representing a promising immunoregulation strategy for maintaining intestinal health in weaned piglets.
Collapse
Affiliation(s)
- Kuanmin M Yang
- State Key Laboratory of Livestock and Poultry Breeding, Key Laboratory of Animal Nutrition and Feed in South China, Ministry of Agriculture and Rural Affairs, Guangdong, China. Key Laboratory of Animal Breeding and Nutrition, Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Institute of Animal Science, China, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, P.R. China
| | - Cui Zhu
- School of Life Sciences and Engineering, Foshan University, Foshan 528225, P.R. China
| | - Li Wang
- State Key Laboratory of Livestock and Poultry Breeding, Key Laboratory of Animal Nutrition and Feed in South China, Ministry of Agriculture and Rural Affairs, Guangdong, China. Key Laboratory of Animal Breeding and Nutrition, Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Institute of Animal Science, China, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, P.R. China
| | - Shuting T Cao
- State Key Laboratory of Livestock and Poultry Breeding, Key Laboratory of Animal Nutrition and Feed in South China, Ministry of Agriculture and Rural Affairs, Guangdong, China. Key Laboratory of Animal Breeding and Nutrition, Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Institute of Animal Science, China, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, P.R. China
| | - Xuefen F Yang
- State Key Laboratory of Livestock and Poultry Breeding, Key Laboratory of Animal Nutrition and Feed in South China, Ministry of Agriculture and Rural Affairs, Guangdong, China. Key Laboratory of Animal Breeding and Nutrition, Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Institute of Animal Science, China, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, P.R. China
| | - Kaiguo G Gao
- State Key Laboratory of Livestock and Poultry Breeding, Key Laboratory of Animal Nutrition and Feed in South China, Ministry of Agriculture and Rural Affairs, Guangdong, China. Key Laboratory of Animal Breeding and Nutrition, Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Institute of Animal Science, China, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, P.R. China
| | - Zongyong Y Jiang
- State Key Laboratory of Livestock and Poultry Breeding, Key Laboratory of Animal Nutrition and Feed in South China, Ministry of Agriculture and Rural Affairs, Guangdong, China. Key Laboratory of Animal Breeding and Nutrition, Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Institute of Animal Science, China, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, P.R. China
| |
Collapse
|
29
|
Metabolomic Profile of Weaned Pigs Challenged with E. coli and Supplemented with Carbadox or Bacillus subtilis. Metabolites 2021; 11:metabo11020081. [PMID: 33573321 PMCID: PMC7911053 DOI: 10.3390/metabo11020081] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/27/2021] [Accepted: 01/28/2021] [Indexed: 11/17/2022] Open
Abstract
This study explored the metabolomic profiles in ileal mucosa and colon digesta in response to enterotoxigenic Escherichia coli F18 (ETEC) infection and dietary use of probiotics and low-dose antibiotics. Weaned pigs (n = 48, 6.17 ± 0.36 kg body weight) were randomly allotted to one of four treatments. Pigs in the negative control (NC) were fed a basal diet without ETEC challenge, whereas pigs in the positive control (PC), antibiotic, and probiotic groups were fed the basal diet, basal diet supplemented with 50 mg/kg of carbadox, or 500 mg/kg of Bacillus subtilis, respectively, and orally challenged with ETEC F18. All pigs were euthanized at day 21 post-inoculation to collect ileal mucosa and colon digesta for untargeted metabolomic profiling using gas chromatography coupled with time-of-flight mass spectrometry. Multivariate analysis highlighted a more distinct metabolomic profile of ileal mucosa metabolites in NC compared to the ETEC-challenged groups. The relative abundance of 19 metabolites from the ileal mucosa including polyamine, nucleotide, monosaccharides, fatty acids, and organic acids was significantly different between the NC and PC groups (q < 0.1). In colon digesta, differential metabolites including 2-monoolein, lactic acid, and maltose were reduced in the carbadox group compared with the probiotics group. In conclusion, several differential metabolites and metabolic pathways were identified in ileal mucosa, which may suggest an ongoing intestinal mucosal repair in the ileum of ETEC-challenged pigs on day 21 post-inoculation.
Collapse
|
30
|
Zong Q, Jing P, Sun S, Wang H, Wu S, Bao W. Effects of HSP27 gene expression on the resistance to Escherichia coli infection in piglets. Gene 2021; 773:145415. [PMID: 33444678 DOI: 10.1016/j.gene.2021.145415] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 12/05/2020] [Accepted: 01/05/2021] [Indexed: 12/29/2022]
Abstract
Heat shock protein 27 (HSP27) plays an important role in protecting cells from various stress factors. This study aimed to investigate the function of HSP27 gene and its regulatory mechanism as infected by Escherichia coli (E. coli) at the tissue and cellular levels. Real-time PCR was used to detect the differential expression of HSP27 gene in F18 resistant and sensitive Sutai pigs and the differential expression upon E. coli F18ab, F18ac, K88ac bacterial supernatant, thallus infection and LPS induction in IPEC-J2. In addition, the HSP27 gene overexpression vector was constructed to detect the effect of the HSP27 gene overexpression on the adhesion of E. coli F18 to IPEC-J2, secretion of pro-inflammatory factors, and the expression of the upstream key genes in Mitogen-activated protein kinase (MAPK) pathway. Ribosomal S6 kinase (RSK2) is an important protein in the MAPK pathway. Therefore, the RSK2 gene overexpression vector was constructed and the number of colonies was counted after co-transfection of HSP27 and RSK2 gene. Results revealed that the expression level of HSP27 gene in resistant individuals in 11 tissues was higher than sensitive type. At the cellular level, the relative expression levels of HSP27 gene were increased after F18ab, F18ac bacterial supernatant, F18ab thallus infection, and LPS induction for 4 h (P < 0.01). The adhesion ability of E. coli F18ab to IPEC-J2 was significantly reduced after HSP27 gene overexpression (P < 0.01), and the concentration of pro-inflammatory factors in the HSP27 gene overexpression group was significantly reduced compared with the control group after F18ab infection (P < 0.05). Furthermore, the expression of RSK2 was significantly increased in HSP27 overexpression group upon F18ab infection (P < 0.01). The colonies quantitative results also showed that the number of colonies was significantly reduced after co-transfection of HSP27 and RSK2 gene. We indicated that the high expression of HSP27 gene may resist the inflammatory response caused by exogenous stress and enhance the ability of IPEC-J2 to resist E. coli F18 infection. RSK2 gene in the MAPK pathway may cooperate with HSP27 gene to participate in the immune response of the organism, which provides a theoretical basis for the study of the mechanism of anti-E. coli infection in piglets.
Collapse
Affiliation(s)
- Qiufang Zong
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, PR China
| | - Pengfei Jing
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, PR China
| | - Shouyong Sun
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, PR China
| | - Haifei Wang
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, PR China; Joint International Research Laboratory of Agriculture & Agri-Product Safety, Yangzhou University, Yangzhou, Jiangsu, PR China
| | - Shenglong Wu
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, PR China; Joint International Research Laboratory of Agriculture & Agri-Product Safety, Yangzhou University, Yangzhou, Jiangsu, PR China
| | - Wenbin Bao
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, PR China; Joint International Research Laboratory of Agriculture & Agri-Product Safety, Yangzhou University, Yangzhou, Jiangsu, PR China.
| |
Collapse
|
31
|
Yang J, Qiu Y, Hu S, Zhu C, Wang L, Wen X, Yang X, Jiang Z. Lactobacillus plantarum inhibited the inflammatory response induced by enterotoxigenic Escherichia coli K88 via modulating MAPK and NF-κB signalling in intestinal porcine epithelial cells. J Appl Microbiol 2020; 130:1684-1694. [PMID: 32870564 DOI: 10.1111/jam.14835] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 07/28/2020] [Accepted: 08/21/2020] [Indexed: 12/22/2022]
Abstract
AIMS To investigate the effects of Lactobacillus plantarum on inflammatory responses induced by ETEC K88 and explore the underlying molecular mechanisms. METHODS AND RESULTS Intestinal porcine cells (IPEC-1) were incubated with 0 or 1 × 108 CFU per well L. plantarum for 4 h, and then these cells were challenged with 0 or 1 × 108 CFU per well ETEC K88 for 2 h. The results showed that pre-treatment of IPEC-1 cells with L. plantarum prevented the increases in the transcript abundance of interleukin-1α (IL-1α), interleukin-6 (IL-6), interleukin-8 (IL-8) and tumour necrosis factor-α (TNF-α) (P < 0·05) caused by ETEC K88. Additionally, L. plantarum inhibited the reduction in peroxisome proliferator-activated receptor-γ (PPAR-γ) expression caused by ETEC K88 (P < 0·05). Moreover, L. plantarum pre-treatment downregulated the phosphorylation levels of c-Jun N-terminal kinase (JNK), extracellular regulated protein kinases 1 and 2 (ERK1/2) and p38 and the nuclear concentration of nuclear factor kappa B p65 (NF-κB p65) (P < 0·05) compared with ETEC K88 group. Silencing experiment further supported that the protective effect of L. plantarum P might mediated by suppression of ETEC-provoked activation of MAPK and NF-κB signalling pathways. CONCLUSIONS Lactobacillus plantarum inhibited the inflammatory response induced by ETEC K88 in IPEC-1 cells via modulating MAPK and NF-κB signalling. SIGNIFICANCE AND IMPACT OF THE STUDY This study elucidated the underlying mechanism in which probiotics protect against intestinal inflammation caused by ETEC K88.
Collapse
Affiliation(s)
- J Yang
- State Key Laboratory of Livestock and Poultry Breeding, Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangdong Public Laboratory of Animal Breeding and Nutrition; Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Y Qiu
- State Key Laboratory of Livestock and Poultry Breeding, Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangdong Public Laboratory of Animal Breeding and Nutrition; Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - S Hu
- State Key Laboratory of Livestock and Poultry Breeding, Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangdong Public Laboratory of Animal Breeding and Nutrition; Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - C Zhu
- State Key Laboratory of Livestock and Poultry Breeding, Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangdong Public Laboratory of Animal Breeding and Nutrition; Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - L Wang
- State Key Laboratory of Livestock and Poultry Breeding, Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangdong Public Laboratory of Animal Breeding and Nutrition; Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - X Wen
- State Key Laboratory of Livestock and Poultry Breeding, Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangdong Public Laboratory of Animal Breeding and Nutrition; Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - X Yang
- State Key Laboratory of Livestock and Poultry Breeding, Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangdong Public Laboratory of Animal Breeding and Nutrition; Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Z Jiang
- State Key Laboratory of Livestock and Poultry Breeding, Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangdong Public Laboratory of Animal Breeding and Nutrition; Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| |
Collapse
|
32
|
Duarte ME, Tyus J, Kim SW. Synbiotic Effects of Enzyme and Probiotics on Intestinal Health and Growth of Newly Weaned Pigs Challenged With Enterotoxigenic F18 + Escherichia coli. Front Vet Sci 2020; 7:573. [PMID: 33033721 PMCID: PMC7509054 DOI: 10.3389/fvets.2020.00573] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 07/17/2020] [Indexed: 12/17/2022] Open
Abstract
This study aimed to investigate the effect of dietary supplementation with xylanase and probiotics on growth performance and intestinal health of nursery pigs challenged with enterotoxigenic Escherichia coli (ETEC). Sixty-four newly weaned pigs (32 barrows and 32 gilts with 7.9 ± 0.4 kg BW) were allotted in a randomized complete block design (2 × 2 factorial). Two factors were ETEC challenge (oral inoculation of saline solution or E. coli F18+ at 6 × 109 CFU) and synbiotics (none or a combination of xylanase 10,000 XU/kg and Bacillus sp. 2 × 108 CFU/kg). All pigs were fed experimental diets following NRC (2012) in two phases (P1 for 10 d and P2 for 11 d). The ETEC was orally inoculated on d 7 after weaning. Feed intake and BW were measured on d 7, 10, 15, and 20. On d 20, pigs were euthanized to collect samples to measure gut health parameters and microbiome. Synbiotics increased (P < 0.05) ADG in phase 1 and ETEC reduced (P < 0.05) ADG and G:F in the post-challenge period. ETEC increased (P < 0.05) the fecal score of pigs from d 7 to 13; however, synbiotics reduced (P < 0.05) it at d 9 and 11 in challenged pigs. ETEC increased (P < 0.05) mucosal MDA, IL-6, Ki-67+, and crypt depth, whereas synbiotics tended to reduce TNFα (P = 0.093), protein carbonyl (P = 0.065), and IL-6 (P = 0.064); reduced (P < 0.05) crypt depth and Ki-67+; and increased (P < 0.05) villus height. ETEC reduced (P < 0.05) the relative abundance of Bacteroidetes and Firmicutes and increased (P < 0.05) the relative abundance of Proteobacteria. In conclusion, ETEC challenge reduced growth performance by affecting microbiome, immune response, and oxidative stress in the jejunum. Synbiotics enhanced growth performance by reducing diarrhea, immune response, and oxidative stress in the jejunum.
Collapse
Affiliation(s)
- Marcos Elias Duarte
- Department of Animal Science, North Carolina State University, Raleigh, NC, United States
| | - James Tyus
- BioResource International, Inc., Durham, NC, United States
| | - Sung Woo Kim
- Department of Animal Science, North Carolina State University, Raleigh, NC, United States
| |
Collapse
|
33
|
Wang W, Liu Y, Tang H, Yu Y, Zhang Q. ITGB5 Plays a Key Role in Escherichia coli F4ac-Induced Diarrhea in Piglets. Front Immunol 2019; 10:2834. [PMID: 31921118 PMCID: PMC6927286 DOI: 10.3389/fimmu.2019.02834] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 11/18/2019] [Indexed: 11/13/2022] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) that expresses F4ac fimbriae is the major pathogenic microorganism responsible for bacterial diarrhea in neonatal piglets. The susceptibility of piglets to ETEC F4ac is determined by a specific receptor on the small intestinal epithelium surface. We performed an iTRAQ-labeled quantitative proteome analysis using a case-control design in which susceptible and resistant full-sib piglets were compared for the protein expression levels. Two thousand two hundred forty-nine proteins were identified, of which 245 were differentially expressed (fold change > 1.5, FDR-adjusted P < 0.05). The differentially expressed proteins fell into four functional classes: (I) cellular adhesion and binding, (II) metabolic process, (III) apoptosis and proliferation, and (IV) immune response. The integrin signaling pathway merited particular interest based on a pathway analysis using statistical overexpression and enrichment tests. Genomic locations of the integrin family genes were determined based on the most recent porcine genome sequence assembly (Sscrofa11.1). Only one gene, ITGB5, which encodes the integrin β5 subunit that assorts with the αv subunit to generate integrin αvβ5, was located within the SSC13q41 region between 13:133161078 and 13:139609422, where strong associations of markers with the ETEC F4ac susceptibility were found in our previous GWAS results. To identify whether integrin αvβ5 is the ETEC F4acR, we established an experimental model for bacterial adhesion using IPEC-J2 cells. Then, the ITGB5 gene was knocked out in IPEC-J2 cell lines using CRISPR/Cas9, resulting in a biallelic deletion cell line (ITGB5 -/-). Disruption of ITGB5 significantly reduced ETEC F4ac adhesion to porcine intestinal epithelial cells. In contrast, overexpression of ITGB5 significantly enhanced the adhesion. A GST pull-down assay with purified FaeG and ITGB5 also showed that FaeG binds directly to ITGB5. Together, the results suggested that ITGB5 is a key factor affecting the susceptibility of piglets to ETEC F4ac.
Collapse
Affiliation(s)
- Wenwen Wang
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Tai'an, China
| | - Yang Liu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Hui Tang
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Tai'an, China
| | - Ying Yu
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Qin Zhang
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Tai'an, China
| |
Collapse
|
34
|
Invited review: Tannins as a potential alternative to antibiotics to prevent coliform diarrhea in weaned pigs. Animal 2019; 14:95-107. [PMID: 31571564 DOI: 10.1017/s1751731119002143] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
In addition to a multifactorial etiology of nutritional, social and environmental stressors, post-weaning diarrhea (PWD) in pigs is often related to infection with specific pathogens such as enterotoxigenic Escherichia coli (ETEC). In swine farming operations, the incidence of PWD is a global concern and is associated with an unbalanced gut status, resulting in poor performance and high antimicrobial consumption via prophylaxis and metaphylaxis. Increases in antimicrobial resistance are reinforcing an already-urgent need for sustainable, alternative solutions for maintaining optimal gut health in livestock. Tannin-rich plants and extracts contain bioactive compounds that could be of great interest in this respect. This review describes how the use of tannins around weaning could be beneficial for pigs, with special emphasis on the reduction of ETEC-related PWD. An overview of the broad chemical diversity of tannins is presented together with their physicochemical and biological properties, as well as how they may be metabolized in the digestive tract. The pharmacological effects exerted by tannins are summarized; more precisely, the possible mechanisms by which tannins can disrupt the different steps of the pathogenesis of ETEC-related PWD are highlighted. The factors affecting the bioactivity of tannins are also discussed, shedding light on the importance of chemical structure among different tannins.
Collapse
|
35
|
Dai C, Yang L, Jin J, Wang H, Wu S, Bao W. Regulation and Molecular Mechanism of TLR5 on Resistance to Escherichia coli F18 in Weaned Piglets. Animals (Basel) 2019; 9:ani9100735. [PMID: 31569693 PMCID: PMC6827021 DOI: 10.3390/ani9100735] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 09/25/2019] [Accepted: 09/26/2019] [Indexed: 12/11/2022] Open
Abstract
Toll-like receptor 5 (TLR5) plays an important role in immune system. In this study, we performed transcriptome analysis of the duodenum in E. coli F18-resistant and -sensitive Sutai weaned piglets and analyzed the differential expression of TLR5. The cellular localization of TLR5 was investigated, and the effect of TLR5 expression on E. coli invasion was evaluated after pig small intestinal epithelial cell lines (IPEC-J2) were stimulated by E. coli. The results showed that TLR5 expression level in duodenum and jejunum were significantly higher in E. coli F18-sensitive than in E. coli F18-resistant piglets. TLR5 protein was mainly expressed in the cytoplasm and cell membrane. The expression of genes associated with the TLR5 signaling pathway were significantly higher in TLR5-overexpressed cells than in control cells. Bacterial adhesion was higher in TLR5-overexpressed cells than in blank cells and lower in TLR5 interference than in blank cells. The core promoter region of TLR5 included two CpG islands and 16 acting elements. The methylation of the mC-6 site in the second CpG island of the promoter region had a regulatory effect on TLR5 expression. Therefore, TLR5 plays an important regulatory role on E. coli invasion. Low expression of TLR5 inhibited the immune response and decreased cell damage, which was conducive to the resistance to E. coli stimulation. In conclusion, this study preliminarily revealed the molecular mechanism of TLR5 gene regulating the resistance of piglets to Escherichia coli, and provided a new candidate gene for screening Escherichia coli resistance markers in pigs.
Collapse
Affiliation(s)
- Chaohui Dai
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China.
| | - Li Yang
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China.
| | - Jian Jin
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China.
| | - Haifei Wang
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China.
| | - Shenglong Wu
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China.
- Joint International Research Laboratory of Agriculture & Agri-Product Safety, Yangzhou University, Yangzhou 225009, China.
| | - Wenbin Bao
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China.
- Joint International Research Laboratory of Agriculture & Agri-Product Safety, Yangzhou University, Yangzhou 225009, China.
| |
Collapse
|
36
|
Luo Y, Xu J, Zhang C, Jiang C, Ma Y, He H, Wu Y, Devriendt B, Cox E, Zhang H. Toll-like receptor 5-mediated IL-17C expression in intestinal epithelial cells enhances epithelial host defense against F4 + ETEC infection. Vet Res 2019; 50:48. [PMID: 31221216 PMCID: PMC6584996 DOI: 10.1186/s13567-019-0665-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 05/27/2019] [Indexed: 12/23/2022] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) are an important cause of post-weaning diarrhea (PWD) in piglets. The IL-17 cytokine family is well known to play important roles in the host defense against bacterial infections at the mucosa. Previously, we reported the potential role of IL-17A in clearing an ETEC infection in piglets. IL-17C, another member of the IL-17 family, is highly expressed in the intestinal epithelium, however, its role during an ETEC infection is still unclear. In this study, we demonstrate that F4+ ETEC induce IL-17C mRNA and protein expression in intestinal tissues as well as in porcine intestinal epithelial cells (IPEC-J2). This IL-17C production is largely dependent on TLR5 signaling in IPEC-J2 cells. Both F4+ ETEC infection and exogenous IL-17C increased the expression of antimicrobial peptides and tight junction proteins, such as porcine beta-defensin (pBD)-2, claudin-1, claudin-2 and occludin in IPEC-J2 cells. Taken together, our data demonstrate that TLR5-mediated IL-17C expression in intestinal epithelial cells enhances mucosal host defense responses in a unique autocrine/paracrine manner in the intestinal epithelium against ETEC infection.
Collapse
Affiliation(s)
- Yu Luo
- Animal Medical Testing Center, Department of Animal Production, Faculty of Agricultural & Biological Engineering, Jinhua Polytechnic, Jinhua, China.
| | - Jia Xu
- Animal Medical Testing Center, Department of Animal Production, Faculty of Agricultural & Biological Engineering, Jinhua Polytechnic, Jinhua, China
| | - Chaoying Zhang
- Animal Medical Testing Center, Department of Animal Production, Faculty of Agricultural & Biological Engineering, Jinhua Polytechnic, Jinhua, China
| | - Chunyan Jiang
- Animal Medical Testing Center, Department of Animal Production, Faculty of Agricultural & Biological Engineering, Jinhua Polytechnic, Jinhua, China
| | - Yanfeng Ma
- Animal Medical Testing Center, Department of Animal Production, Faculty of Agricultural & Biological Engineering, Jinhua Polytechnic, Jinhua, China
| | - Haijian He
- Animal Medical Testing Center, Department of Animal Production, Faculty of Agricultural & Biological Engineering, Jinhua Polytechnic, Jinhua, China
| | - Yuan Wu
- Animal Medical Testing Center, Department of Animal Production, Faculty of Agricultural & Biological Engineering, Jinhua Polytechnic, Jinhua, China
| | - Bert Devriendt
- Laboratory of Immunology, Faculty of Veterinary Medicine, Ghent University, Ghent, Belgium
| | - Eric Cox
- Laboratory of Immunology, Faculty of Veterinary Medicine, Ghent University, Ghent, Belgium
| | - Hongbin Zhang
- Animal Medical Testing Center, Department of Animal Production, Faculty of Agricultural & Biological Engineering, Jinhua Polytechnic, Jinhua, China
| |
Collapse
|
37
|
Mani S, Toapanta FR, McArthur MA, Qadri F, Svennerholm AM, Devriendt B, Phalipon A, Cohen D, Sztein MB. Role of antigen specific T and B cells in systemic and mucosal immune responses in ETEC and Shigella infections, and their potential to serve as correlates of protection in vaccine development. Vaccine 2019; 37:4787-4793. [PMID: 31230883 PMCID: PMC7413037 DOI: 10.1016/j.vaccine.2019.03.040] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 03/07/2019] [Accepted: 03/19/2019] [Indexed: 12/13/2022]
Abstract
The generation of robust systemic and mucosal antibody and cell-mediated immune (CMI) responses that are protective, long-lasting, and can quickly be recalled upon subsequent re-exposure to the cognate antigen is the key to the development of effective vaccine candidates. These responses, whether they represent mechanistic or non-mechanistic immunological correlates of protection, usually entail the activation of T cell memory and effector subsets (T-CMI) and induction of long-lasting memory B cells. However, for ETEC and Shigella, the precise role of these key immune cells in primary and secondary (anamnestic) immune responses remains ill-defined. A workshop to address immune correlates for ETEC and Shigella, in general, and to elucidate the mechanistic role of T-cell subsets and B-cells, both systemically and in the mucosal microenvironment, in the development of durable protective immunity against ETEC and Shigella was held at the recent 2nd Vaccines against Shigella and ETEC (VASE) conference in June 2018. This report is a summary of the presentations and the discussion that ensued at the workshop.
Collapse
Affiliation(s)
| | - Franklin R Toapanta
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Monica A McArthur
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Firdausi Qadri
- Infectious Diseases Division, International Center for Diarrheal Diseases Research, Dhaka, Bangladesh
| | - Ann-Mari Svennerholm
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Sweden
| | - Bert Devriendt
- Faculty of Veterinary Medicine, Department of Virology, Parasitology, and Immunology, Ghent University, Belgium
| | - Armelle Phalipon
- Molecular Microbial Pathogenesis, INSERM U1202, Institut Pasteur, Paris, France
| | - Daniel Cohen
- School of Public Health, Sackler Faculty of Medicine, Tel Aviv University, Israel
| | - Marcelo B Sztein
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States.
| |
Collapse
|
38
|
Qiu Y, Yang X, Wang L, Gao K, Jiang Z. L-Arginine Inhibited Inflammatory Response and Oxidative Stress Induced by Lipopolysaccharide via Arginase-1 Signaling in IPEC-J2 Cells. Int J Mol Sci 2019; 20:ijms20071800. [PMID: 30979040 PMCID: PMC6479672 DOI: 10.3390/ijms20071800] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 04/05/2019] [Accepted: 04/09/2019] [Indexed: 12/18/2022] Open
Abstract
This study aimed to explore the effect of L-arginine on lipopolysaccharide (LPS)-induced inflammatory response and oxidative stress in IPEC-2 cells. We found that the expression of toll-like receptor 4 (TLR4), myeloid differentiation primary response 88 (MyD88), cluster of differentiation 14 (CD14), nuclear factor-kappaBp65 (NF-κBp65), chemokine-8 (IL-8), tumor necrosis factor (TNF-α) and chemokine-6 (IL-6) mRNA were significantly increased by LPS. Exposure to LPS induced oxidative stress as reactive oxygen species (ROS) and malonaldehyde (MDA) production were increased while glutathione peroxidase (GSH-Px) were decreased in LPS-treated cells compared to those in the control. LPS administration also effectively induced cell growth inhibition through induction of G0/G1 cell cycle arrest. However, compared with the LPS group, cells co-treatment with L-arginine effectively increased cell viability and promoted the cell cycle into the S phase; L-arginine exhibited an anti-inflammatory effect in alleviating inflammation induced by LPS by reducing the abundance of TLR4, MyD88, CD14, NF-κBp65, and IL-8 transcripts. Cells treated with LPS+L-arginine significantly enhanced the content of GSH-Px, while they decreased the production of ROS and MDA compared with the LPS group. Furthermore, L-arginine increased the activity of arginase-1 (Arg-1), while Arg-1 inhibitor abolished the protection of arginine against LPS-induced inflammation and oxidative stress. Taken together, these results suggested that L-arginine exerted its anti-inflammatory and antioxidant effects to protect IPEC-J2 cells from inflammatory response and oxidative stress challenged by LPS at least partly via the Arg-1 signaling pathway.
Collapse
Affiliation(s)
- Yueqin Qiu
- State Key Laboratory of Livestock and Poultry Breeding; Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture; Guangdong Public Laboratory of Animal Breeding and Nutrition; Guangdong Key Laboratory of Animal Breeding and Nutrition; Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China.
- College of Animal Science, South China Agricultural University, Guangzhou 510642, China.
| | - Xuefen Yang
- State Key Laboratory of Livestock and Poultry Breeding; Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture; Guangdong Public Laboratory of Animal Breeding and Nutrition; Guangdong Key Laboratory of Animal Breeding and Nutrition; Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China.
| | - Li Wang
- State Key Laboratory of Livestock and Poultry Breeding; Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture; Guangdong Public Laboratory of Animal Breeding and Nutrition; Guangdong Key Laboratory of Animal Breeding and Nutrition; Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China.
| | - Kaiguo Gao
- State Key Laboratory of Livestock and Poultry Breeding; Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture; Guangdong Public Laboratory of Animal Breeding and Nutrition; Guangdong Key Laboratory of Animal Breeding and Nutrition; Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China.
| | - Zongyong Jiang
- State Key Laboratory of Livestock and Poultry Breeding; Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture; Guangdong Public Laboratory of Animal Breeding and Nutrition; Guangdong Key Laboratory of Animal Breeding and Nutrition; Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China.
| |
Collapse
|
39
|
Effect of Puerarin, Baicalin and Berberine Hydrochloride on the Regulation of IPEC-J2 Cells Infected with Enterotoxigenic Escherichia coli. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 2019:7438593. [PMID: 30891078 PMCID: PMC6390247 DOI: 10.1155/2019/7438593] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 07/06/2018] [Accepted: 01/17/2019] [Indexed: 11/17/2022]
Abstract
Puerarin, baicalin and berberine hydrochloride are the main components of Gegen Qinlian Decoction, which has been used to treat diarrhoea in China for hundreds of years, yet the biological function and molecular mechanism of these components are not clear. To investigate the effects of puerarin, baicalin, and berberine hydrochloride on the regulation of porcine intestinal epithelial cells (IPEC-J2 cells) infected with enterotoxigenic Escherichia coli (ETEC). IPEC-J2 cells were pretreated with puerarin (200 μg/mL), baicalin (1 μg/mL), and berberine hydrochloride (100 μg/mL) at 37°C for 3 h and then coincubated with the F4ac ETEC bacterial strain 200 at 37°C for 3 h. ETEC infection damaged the structure of IPEC-J2 cells, upregulated mucin 4 (P < 0.01) and mucin 13 mRNA (P < 0.05) expression, increased the apoptosis rate (P < 0.05), and promoted inflammatory responses (IL-6 and CXCL-2 mRNA expression) in IPEC-J2 cells by activating the nuclear factor-κB (NF-κB) signaling pathway. Pretreatment with puerarin, baicalin, and berberine hydrochloride improved the structure and morphology of IPEC-J2 cells and inhibited ETEC adhesion by downregulating specific adhesion molecules. Pretreatment with baicalin decreased the inflammatory response; pretreatment with baicalin and berberine hydrochloride decreased the inflammatory response mediated by the NF-κB signaling pathway. Pretreatment with puerarin, baicalin, and berberine hydrochloride protected IPEC-J2 cells from ETEC infection by inhibiting bacterial adhesion and inflammatory responses.
Collapse
|
40
|
Xia L, Dai L, Zhu L, Hu W, Yang Q. Proteomic Analysis of IPEC-J2 Cells in Response to Coinfection by Porcine Transmissible Gastroenteritis Virus and Enterotoxigenic Escherichia coli K88. Proteomics Clin Appl 2018; 11. [PMID: 29090858 DOI: 10.1002/prca.201600137] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 09/22/2017] [Indexed: 01/03/2023]
Abstract
SCOPE Piglet diarrhea causes large economic losses to the swine industry. Epidemiological investigations show that piglet diarrhea is often caused by mixed infections, but the mechanisms by which multiple microorganisms cause disease are unclear. EXPERIMENTAL DESIGN Because transmissible gastroenteritis virus (TGEV) and enterotoxigenic Escherichia coli K88 (ETEC K88) are important contributors to piglet diarrhea, coinfection experiments are conducted using porcine intestinal columnar epithelial cells (IPEC-J2) as a model system. In order to evaluate piglet diarrhea caused TGEV and ETEC K88, the authors examin the effects of coinfection in IPEC-J2 cells. In TGEV pre-infected IPEC-J2 cells, ETEC K88 adhesion is enhanced over uninfected cells. ETEC K88 is also found to inhibit the proliferation of TGEV. Additionally, cytokine levels (IL-1β, IL-6, IL-8, and TNF-α) in coinfected cells are lower than cells infected by TGEV alone, and higher than cells infected by ETEC K88 alone. LCMS/MS coupled to isobaric tags for relative and absolute quantification (iTRAQ) is used to profile expressed proteins in IPEC-J2 cells infected by TGEV alone, ETEC K88 alone, and by both agents together. RESULTS 77, 89, and 136 differentially expressed proteins are identified in TGEV infected, ETEC K88 infected, and coinfected cells, respectively. CONCLUSION AND CLINICAL RELEVANCE Based on these data, the authors suspect that integrin α5 might enable TGEV to promote ETEC K88 adhesion. This study is the first to analyze piglet diarrhea caused by TGEV-ETEC K88 coinfection using high-throughput quantitative proteomics. The results advance the understanding of coinfection and its role in causing piglet diarrhea.
Collapse
Affiliation(s)
- Lu Xia
- College of veterinary medicine, Nanjing Agricultural University, Jiangsu, PR China
| | - Lei Dai
- College of veterinary medicine, Nanjing Agricultural University, Jiangsu, PR China
| | - Liqi Zhu
- College of veterinary medicine, Nanjing Agricultural University, Jiangsu, PR China
| | - Weiwei Hu
- College of veterinary medicine, Nanjing Agricultural University, Jiangsu, PR China
| | - Qian Yang
- College of veterinary medicine, Nanjing Agricultural University, Jiangsu, PR China
| |
Collapse
|
41
|
Hoelzer K, Bielke L, Blake DP, Cox E, Cutting SM, Devriendt B, Erlacher-Vindel E, Goossens E, Karaca K, Lemiere S, Metzner M, Raicek M, Collell Suriñach M, Wong NM, Gay C, Van Immerseel F. Vaccines as alternatives to antibiotics for food producing animals. Part 2: new approaches and potential solutions. Vet Res 2018; 49:70. [PMID: 30060759 PMCID: PMC6066917 DOI: 10.1186/s13567-018-0561-7] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 12/22/2017] [Indexed: 12/22/2022] Open
Abstract
Vaccines and other alternative products are central to the future success of animal agriculture because they can help minimize the need for antibiotics by preventing and controlling infectious diseases in animal populations. To assess scientific advancements related to alternatives to antibiotics and provide actionable strategies to support their development, the United States Department of Agriculture, with support from the World Organisation for Animal Health, organized the second International Symposium on Alternatives to Antibiotics. It focused on six key areas: vaccines; microbial-derived products; non-nutritive phytochemicals; immune-related products; chemicals, enzymes, and innovative drugs; and regulatory pathways to enable the development and licensure of alternatives to antibiotics. This article, the second part in a two-part series, highlights new approaches and potential solutions for the development of vaccines as alternatives to antibiotics in food producing animals; opportunities, challenges and needs for the development of such vaccines are discussed in the first part of this series. As discussed in part 1 of this manuscript, many current vaccines fall short of ideal vaccines in one or more respects. Promising breakthroughs to overcome these limitations include new biotechnology techniques, new oral vaccine approaches, novel adjuvants, new delivery strategies based on bacterial spores, and live recombinant vectors; they also include new vaccination strategies in-ovo, and strategies that simultaneously protect against multiple pathogens. However, translating this research into commercial vaccines that effectively reduce the need for antibiotics will require close collaboration among stakeholders, for instance through public–private partnerships. Targeted research and development investments and concerted efforts by all affected are needed to realize the potential of vaccines to improve animal health, safeguard agricultural productivity, and reduce antibiotic consumption and resulting resistance risks.
Collapse
Affiliation(s)
- Karin Hoelzer
- The Pew Charitable Trusts, 901 E Street NW, Washington, DC, 20004, USA.
| | - Lisa Bielke
- Ohio Agriculture and Research Development Center, Animal Sciences, Ohio State University, 202 Gerlaugh Hall, 1680 Madison Ave., Wooster, OH, 44691, USA
| | - Damer P Blake
- Pathobiology and Population Sciences, Royal Veterinary College, University of London, Hawkshead Lane, North Mymms, Hertfordshire, AL9 7TA, UK
| | - Eric Cox
- Department of Virology, Parasitology and Immunology, Faculty of Veterinary Medicine, Ghent University, Salsiburylaan 133, 9820, Merelbeke, Belgium
| | - Simon M Cutting
- School of Biological Sciences, Royal Holloway University of London, Egham, Surrey, TW20 0EX, UK
| | - Bert Devriendt
- Department of Virology, Parasitology and Immunology, Faculty of Veterinary Medicine, Ghent University, Salsiburylaan 133, 9820, Merelbeke, Belgium
| | - Elisabeth Erlacher-Vindel
- Science and New Technologies Department, World Organisation for Animal Health (OIE), 12 Rue de Prony, 75017, Paris, France
| | - Evy Goossens
- Department of Pathology, Bacteriology and Avian Diseases, Faculty of Veterinary Medicine, Ghent University, Salsiburylaan 133, 9820, Merelbeke, Belgium
| | - Kemal Karaca
- Elanco Animal Health, 2500 Innovation Way, Greenfield, IN, USA
| | | | - Martin Metzner
- RIPAC-LABOR GmbH, Am Mühlenberg 11, 14476, Potsdam, Germany
| | - Margot Raicek
- Science and New Technologies Department, World Organisation for Animal Health (OIE), 12 Rue de Prony, 75017, Paris, France
| | | | - Nora M Wong
- The Pew Charitable Trusts, 901 E Street NW, Washington, DC, 20004, USA
| | - Cyril Gay
- Office of National Programs, Agricultural Research Service, USDA, Sunnyside Ave, 5601, Beltsville, MD, USA
| | - Filip Van Immerseel
- Department of Pathology, Bacteriology and Avian Diseases, Faculty of Veterinary Medicine, Ghent University, Salsiburylaan 133, 9820, Merelbeke, Belgium
| |
Collapse
|
42
|
Enhancement of immune response of piglets to PCV-2 vaccine by porcine IL-2 and fusion IL-4/6 gene entrapped in chitosan nanoparticles. Res Vet Sci 2018; 117:224-232. [DOI: 10.1016/j.rvsc.2017.12.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2017] [Revised: 11/17/2017] [Accepted: 12/14/2017] [Indexed: 12/20/2022]
|
43
|
Binding determinants in the interplay between porcine aminopeptidase N and enterotoxigenic Escherichia coli F4 fimbriae. Vet Res 2018; 49:23. [PMID: 29482635 PMCID: PMC5828407 DOI: 10.1186/s13567-018-0519-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 02/06/2018] [Indexed: 11/25/2022] Open
Abstract
The binding of F4+ enterotoxigenic Escherichia coli (ETEC) and the specific receptor on porcine intestinal epithelial cells is the initial step in F4+ ETEC infection. Porcine aminopeptidase N (APN) is a newly discovered receptor for F4 fimbriae that binds directly to FaeG adhesin, which is the major subunit of the F4 fimbriae variants F4ab, F4ac, and F4ad. We used overlapping peptide assays to map the APN-FaeG binding sites, which has facilitated in the identifying the APN-binding amino acids that are located in the same region of FaeG variants, thereby limiting the major binding regions of APN to 13 peptides. To determine the core sequence motif, a panel of FaeG peptides with point mutations and FaeG mutants were constructed. Pull-down and binding reactivity assays using piglet intestines determined that the amino acids G159 of F4ab, N209 and L212 of F4ac, and A200 of F4ad were the critical residues for APN binding of FaeG. We further show using ELISA and confocal microscopy assay that amino acids 553–568, and 652–670 of the APN comprise the linear epitope for FaeG binding in all three F4 fimbriae variants.
Collapse
|
44
|
Dubreuil JD. Enterotoxigenic Escherichia coli and probiotics in swine: what the bleep do we know? BIOSCIENCE OF MICROBIOTA, FOOD AND HEALTH 2017; 36:75-90. [PMID: 28785529 PMCID: PMC5510153 DOI: 10.12938/bmfh.16-030] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 04/18/2017] [Indexed: 12/28/2022]
Abstract
The concept of certain microorganisms conferring direct benefits to the host relates to the term "probiotic". Probiotics are microorganisms, bacteria, or yeast that when administered orally in sufficient quantity can counteract the effect of pathogenic microorganisms. The gastrointestinal (GI) tract is the site where probiotics are believed to play the most important role. The proposed effects of probiotics include antagonism of pathogens, interference with adherence, competition for nutrients, enterotoxin inactivation, modulation of the immune response, and strengthening of the intestinal barrier. From birth to postweaning, piglets are very sensitive to gut colonisation by pathogens. Enterotoxigenic Escherichia coli represents one of the most common agents of swine diarrhoea. The enterotoxins produced by this E. coli virotype are responsible for the loss of electrolytes and water observed following infection. This review addresses more specifically the studies done during the last 10 years deciphering the molecular mechanisms at play between host cell and probiotic interactions in the swine GI tract.
Collapse
Affiliation(s)
- Jean Daniel Dubreuil
- Department of Pathology and Microbiology, Faculté de Médecine Vétérinaire, Université de Montréal, 3200 rue Sicotte, Saint-Hyacinthe, Québec J2S 7C6, Canada
| |
Collapse
|
45
|
Tian Z, Liu X, Dai R, Xiao Y, Wang X, Bi D, Shi D. Enterococcus faecium HDRsEf1 Protects the Intestinal Epithelium and Attenuates ETEC-Induced IL-8 Secretion in Enterocytes. Mediators Inflamm 2016; 2016:7474306. [PMID: 27890970 PMCID: PMC5116501 DOI: 10.1155/2016/7474306] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Revised: 08/21/2016] [Accepted: 09/28/2016] [Indexed: 12/25/2022] Open
Abstract
The probiotic Enterococcus faecium HDRsEf1 (Ef1) has been shown to have positive effects on piglet diarrhoea, but the mechanism has not yet been elucidated. In this study, using the IPEC-J2 cell line to mimic intestinal epithelial cells and enterotoxigenic Escherichia coli (ETEC) K88ac as a representative intestinal pathogen, the mechanism underlying Ef1 protection against an enteropathogen was investigated. The results demonstrated that Ef1 was effective in displacing K88ac from the IPEC-J2 cell layer. Moreover, Ef1 and its cell-free supernatant (S-Ef1) modulate IL-8 released by IPEC-J2 cells. Ef1 and its cell-free supernatant showed the potential to protect enterocytes from an acute inflammatory response. In addition, Ef1 and its cell-free supernatant increased the transepithelial electrical resistance (TEER) of the enterocyte monolayer, thus strengthening the intestinal barrier against ETEC. These results may contribute to the development of therapeutic interventions using Ef1 in intestinal disorders of piglets.
Collapse
Affiliation(s)
- Zhongyuan Tian
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Xiaofang Liu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Ran Dai
- College of Animal Science and Technology, Agricultural University of Hebei, Baoding 071000, China
| | - Yuncai Xiao
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Xiliang Wang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Dingren Bi
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Deshi Shi
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| |
Collapse
|
46
|
Dubreuil JD, Isaacson RE, Schifferli DM. Animal Enterotoxigenic Escherichia coli. EcoSal Plus 2016; 7:10.1128/ecosalplus.ESP-0006-2016. [PMID: 27735786 PMCID: PMC5123703 DOI: 10.1128/ecosalplus.esp-0006-2016] [Citation(s) in RCA: 193] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Indexed: 12/13/2022]
Abstract
Enterotoxigenic Escherichia coli (ETEC) is the most common cause of E. coli diarrhea in farm animals. ETEC are characterized by the ability to produce two types of virulence factors: adhesins that promote binding to specific enterocyte receptors for intestinal colonization and enterotoxins responsible for fluid secretion. The best-characterized adhesins are expressed in the context of fimbriae, such as the F4 (also designated K88), F5 (K99), F6 (987P), F17, and F18 fimbriae. Once established in the animal small intestine, ETEC produce enterotoxin(s) that lead to diarrhea. The enterotoxins belong to two major classes: heat-labile toxins that consist of one active and five binding subunits (LT), and heat-stable toxins that are small polypeptides (STa, STb, and EAST1). This review describes the disease and pathogenesis of animal ETEC, the corresponding virulence genes and protein products of these bacteria, their regulation and targets in animal hosts, as well as mechanisms of action. Furthermore, vaccines, inhibitors, probiotics, and the identification of potential new targets by genomics are presented in the context of animal ETEC.
Collapse
Affiliation(s)
- J Daniel Dubreuil
- Faculté de Médecine Vétérinaire, Université de Montréal, Québec J2S 7C6, Canada
| | - Richard E Isaacson
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN 55108
| | - Dieter M Schifferli
- School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104
| |
Collapse
|
47
|
YANG XIANGWU, XIAO ZHIMING, LIU FEN, CHEN SHUAI, TANG WULIANG, ZHANG DECAI, LIU SHAOJUN. Enterotoxigenic Escherichia coli infection alters intestinal immunity in mice. Mol Med Rep 2016; 14:825-30. [DOI: 10.3892/mmr.2016.5302] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 04/07/2016] [Indexed: 11/06/2022] Open
|
48
|
Xia P, Wang Y, Zhu C, Zou Y, Yang Y, Liu W, Hardwidge PR, Zhu G. Porcine aminopeptidase N binds to F4+ enterotoxigenic Escherichia coli fimbriae. Vet Res 2016; 47:24. [PMID: 26857562 PMCID: PMC4746772 DOI: 10.1186/s13567-016-0313-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Accepted: 01/25/2016] [Indexed: 11/26/2022] Open
Abstract
F4+ enterotoxigenic Escherichia coli (ETEC) strains cause diarrheal disease in neonatal and post-weaned piglets. Several different host receptors for F4 fimbriae have been described, with porcine aminopeptidase N (APN) reported most recently. The FaeG subunit is essential for the binding of the three F4 variants to host cells. Here we show in both yeast two-hybrid and pulldown assays that APN binds directly to FaeG, the major subunit of F4 fimbriae, from three serotypes of F4+ ETEC. Modulating APN gene expression in IPEC-J2 cells affected ETEC adherence. Antibodies raised against APN or F4 fimbriae both reduced ETEC adherence. Thus, APN mediates the attachment of F4+E. coli to intestinal epithelial cells.
Collapse
Affiliation(s)
- Pengpeng Xia
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China. .,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China.
| | - Yiting Wang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China. .,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China.
| | - Congrui Zhu
- College of Animal Medicine, Nanjing Agriculture University, Nanjing, 210095, China.
| | - Yajie Zou
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China. .,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China.
| | - Ying Yang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China. .,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China.
| | - Wei Liu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China. .,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China.
| | - Philip R Hardwidge
- College of Veterinary Medicine, Kansas State University, Manhattan, KS, 66506, USA.
| | - Guoqiang Zhu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China. .,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China.
| |
Collapse
|
49
|
Luo Y, Van Nguyen U, de la Fe Rodriguez PY, Devriendt B, Cox E. F4+ ETEC infection and oral immunization with F4 fimbriae elicits an IL-17-dominated immune response. Vet Res 2015; 46:121. [PMID: 26490738 PMCID: PMC4618862 DOI: 10.1186/s13567-015-0264-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 10/02/2015] [Indexed: 02/06/2023] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) are an important cause of post-weaning diarrhea (PWD) in piglets. Porcine-specific ETEC strains possess different fimbrial subtypes of which F4 fimbriae are the most frequently associated with ETEC-induced diarrhea in piglets. These F4 fimbriae are potent oral immunogens that induce protective F4-specific IgA antibody secreting cells at intestinal tissues. Recently, T-helper 17 (Th17) cells have been implicated in the protection of the host against extracellular pathogens. However, it remains unknown if Th17 effector responses are needed to clear ETEC infections. In the present study, we aimed to elucidate if ETEC elicits a Th17 response in piglets and if F4 fimbriae trigger a similar response. F4+ ETEC infection upregulated IL-17A, IL-17F, IL-21 and IL-23p19, but not IL-12 and IFN-γ mRNA expression in the systemic and mucosal immune system. Similarly, oral immunization with F4 fimbriae triggered a Th17 signature evidenced by an upregulated mRNA expression of IL-17F, RORγt, IL-23p19 and IL-21 in the peripheral blood mononuclear cells (PBMCs). Intriguingly, IL-17A mRNA levels were unaltered. To further evaluate this difference between systemic and mucosal immune responses, we assayed the cytokine mRNA profile of F4 fimbriae stimulated PBMCs. F4 fimbriae induced IL-17A, IL-17F, IL-22 and IL-23p19, but downregulated IL-17B mRNA expression. Altogether, these data indicate a Th17 dominated response upon oral immunization with F4 fimbriae and F4+ ETEC infection. Our work also highlights that IL-17B and IL-17F participate in the immune response to protect the host against F4+ ETEC infection and could aid in the design of future ETEC vaccines.
Collapse
Affiliation(s)
- Yu Luo
- Laboratory of Veterinary Immunology, Department of Virology, Parasitology and Immunology, Faculty of Veterinary Medicine, Ghent University, Ghent, Belgium.
| | - Ut Van Nguyen
- Laboratory of Veterinary Immunology, Department of Virology, Parasitology and Immunology, Faculty of Veterinary Medicine, Ghent University, Ghent, Belgium.
| | - Pedro Y de la Fe Rodriguez
- Department of Veterinary Medicine and Zootechnics, Universidad Central "Marta Abreu" de Las Villas, Carretera a Camajuani km 5½, 54830, Santa Clara, Villa Clara, Cuba.
| | - Bert Devriendt
- Laboratory of Veterinary Immunology, Department of Virology, Parasitology and Immunology, Faculty of Veterinary Medicine, Ghent University, Ghent, Belgium.
| | - Eric Cox
- Laboratory of Veterinary Immunology, Department of Virology, Parasitology and Immunology, Faculty of Veterinary Medicine, Ghent University, Ghent, Belgium.
| |
Collapse
|
50
|
Xia P, Song Y, Zou Y, Yang Y, Zhu G. F4+ enterotoxigenic Escherichia coli (ETEC) adhesion mediated by the major fimbrial subunit FaeG. J Basic Microbiol 2015; 55:1118-24. [PMID: 25847483 DOI: 10.1002/jobm.201400901] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 03/13/2015] [Indexed: 11/07/2022]
Abstract
The FaeG subunit is the major constituent of F4(+) fimbriae, associated with glycoprotein and/or glycolipid receptor recognition and majorly contributes to the pathogen attachment to the host cells. To investigate the key factor involved in the fimbrial binding of F4(+) Escherichia coli, both the recombinant E. coli SE5000 strains carrying the fae operon gene clusters that express the different types of fimbriae in vitro, named as rF4ab, rF4ac, and rF4ad, respectively, corresponding to the fimbrial types F4ab, F4ac, and F4ad, and the three isogenic in-frame faeG gene deletion mutants were constructed. The adhesion assays and adhesion inhibition assays showed that ΔfaeG mutants had a significant reduction in the binding to porcine brush border as well as the intestinal epithelial cell lines, while the complemented strain ΔfaeG/pfaeG restored the adhesion function. The recombinant bacterial strains rF4ab, rF4ac, and rF4ad have the same binding property as wild-type F4(+) E. coli strains do and improvement in terms of binding to porcine brush border and the intestinal epithelial cells, and the adherence was blocked by the monoclonal antibody anti-F4 fimbriae. These data demonstrate that the fimbrial binding of F4(+) E. coli is directly mediated by the major FaeG subunit.
Collapse
Affiliation(s)
- Pengpeng Xia
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Yujie Song
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Yajie Zou
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Ying Yang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Guoqiang Zhu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| |
Collapse
|