1
|
Perruchot MH, Boudry G, Mayeur-Nickel F, Grondin M, Wiart-Letort S, Giblin L, Grundy MML. In Vitro Evaluation of Intestinal Barrier Function after Exposure to Digested Pea Ingredients─Food Matrix Effect. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024. [PMID: 39681414 DOI: 10.1021/acs.jafc.4c09963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2024]
Abstract
Dietary fibers (DF) are important components of human and animal diets. However, they can decrease protein digestibility and absorption and thus the nutritional value of a food. The aim of this study was to investigate how the form of delivery of pea DF impacted the integrity of the intestinal barrier and, thereby, the potential absorption of molecules. To this end, two pea flours, with either intact or ruptured cell walls, and two controls, pea fibers and pea protein, were digested in vitro and the digesta obtained applied onto a jejunum porcine cell line (IPEC-J2 cells). Cell viability and integrity were evaluated by transepithelial electrical resistance measurement, colorimetric assay (MTS), and immunohistochemistry for tight junction proteins. Additionally, the diffusion of FITC-dextran (FD4) and lucifer yellow (LY) through the epithelial cell monolayers was monitored. The digested pea samples did not alter the IPEC-J2 viability and permeability. For instance, no difference in the diffusion of molecules either FD4 or LY across the monolayers was observed between the different digesta and the control. Similarly, no effect was observed in ZO-1 labeling intensity compared to the control. This study demonstrated that intestinal integrity was maintained whether pea cell walls were intact or ruptured.
Collapse
Affiliation(s)
| | - Gaëlle Boudry
- NUMECAN, INSERM, INRAE, Université de Rennes, Saint Gilles 35590, France
| | | | | | | | - Linda Giblin
- Teagasc Food Research Centre, Moorepark, Fermoy, Co Cork P61 C996, Ireland
| | | |
Collapse
|
2
|
De Angelis E, Borghetti P, Passeri B, Cavalli V, Ferrari L, Andrani M, Martelli P, Saleri R. Hyperosmotic Stress Induces the Expression of Organic Osmolyte Transporters in Porcine Intestinal Cells and Betaine Exerts a Protective Effect on the Barrier Function. Biomedicines 2024; 12:2391. [PMID: 39457703 PMCID: PMC11503993 DOI: 10.3390/biomedicines12102391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 10/16/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024] Open
Abstract
Background/objectives: The porcine intestinal epithelium plays a fundamental role as a defence interface against pathogens. Its alteration can cause severe inflammatory conditions and diseases. Hyperosmotic stress under physiological conditions and upon pathogen challenge can cause malabsorption. Different cell types counteract the osmolarity increase by accumulating organic osmolytes such as betaine, taurine, and myo-inositol through specific transporters. Betaine is known for protecting cells from hyperosmotic stress and has positive effects when fed to pigs. The aim of this study is to demonstrate the modulation of osmolyte transporters gene expression in IPEC-J2 during osmolarity changes and assess the effects of betaine. Methods: IPEC-J2 were seeded in transwells, where differentiate as a polarized monolayer. Epithelial cell integrity (TEER), oxidative stress (NO) and gene expression of osmolyte transporters, tight junction proteins (TJp) and pro-inflammatory cytokines were evaluated. Results: Cells treated with NaCl hyperosmolar medium (500 mOsm/L) showed a TEER decrease at 3 h and detachment within 24 h, associated with an osmolyte transporters reduction. IPEC-J2 treated with mannitol hyperosmolar medium (500 mOsm/L) upregulated taurine (TauT), myo-inositol (SMIT) and betaine (BGT1) transporters expression. A decrease in TJp expression was associated with a TEER decrease and an increase in TNFα, IL6, and IL8. Betaine could attenuate the hyperosmolarity-induced reduction in TEER and TJp expression, the NO increase and cytokines upregulation. Conclusions: This study demonstrates the expression of osmolyte transporters in IPEC-J2, which was upregulated upon hyperosmotic treatment. Betaine counteracts changes in intracellular osmolarity by contributing to maintaining the epithelial barrier function and reducing the inflammatory condition. Compatible osmolytes may provide beneficial effects in therapies for diseases characterized by inflammation and TJp-related dysfunctions.
Collapse
Affiliation(s)
| | | | | | | | | | - Melania Andrani
- Department of Veterinary Science, University of Parma, Strada del Taglio 10, 43126 Parma, Italy; (E.D.A.); (P.B.); (B.P.); (V.C.); (L.F.); (P.M.); (R.S.)
| | | | | |
Collapse
|
3
|
Binder ARD, Mussack V, Kirchner B, Pfaffl MW. Growth behavior and mRNA expression profiling during growth of IPEC-J2 cells. BMC Res Notes 2024; 17:154. [PMID: 38840260 PMCID: PMC11155027 DOI: 10.1186/s13104-024-06812-w] [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: 03/04/2024] [Accepted: 05/22/2024] [Indexed: 06/07/2024] Open
Abstract
OBJECTIVE The IPEC-J2 cell line is used as an in vitro small intestine model for swine, but it is also used as a model for the human intestine, presenting a relatively unique setting. By combining electric cell-substrate impedance sensing, with next-generation-sequencing technology, we showed that mRNA gene expression profiles and related pathways can depend on the growth phase of IPEC-J2 cells. Our investigative approach welcomes scientists to reproduce or modify our protocols and endorses putting their gene expression data in the context of the respective growth phase of the cells. RESULTS Three time points are presented: (TP1) 1 h after medium change (= 6 h after seeding of cells), (TP2) the time point of the first derivative maximum of the cell growth curve, and a third point at the beginning of the plateau phase (TP3). Significantly outstanding at TP1 compared to TP2 was upregulated PLEKHN1, further FOSB and DEGS2 were significantly downregulated at TP2 compared to TP3. Any provided data can be used to improve next-generation experiments with IPEC-J2 cells.
Collapse
Affiliation(s)
- A Ronja D Binder
- Division of Animal Physiology and Immunology, TUM School of Life Sciences, Technical University of Munich, Weihenstephaner Berg 3, 85354, Freising, Germany.
| | - Veronika Mussack
- Division of Animal Physiology and Immunology, TUM School of Life Sciences, Technical University of Munich, Weihenstephaner Berg 3, 85354, Freising, Germany
| | - Benedikt Kirchner
- Division of Animal Physiology and Immunology, TUM School of Life Sciences, Technical University of Munich, Weihenstephaner Berg 3, 85354, Freising, Germany
| | - Michael W Pfaffl
- Division of Animal Physiology and Immunology, TUM School of Life Sciences, Technical University of Munich, Weihenstephaner Berg 3, 85354, Freising, Germany
| |
Collapse
|
4
|
Andrani M, Ferrari L, Borghetti P, Cavalli V, De Angelis E, Ravanetti F, Dall'Olio E, Martelli P, Saleri R. Short-chain fatty acids modulate the IPEC-J2 cell response to pathogenic E. coli LPS-activated PBMC. Res Vet Sci 2024; 171:105231. [PMID: 38513460 DOI: 10.1016/j.rvsc.2024.105231] [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: 07/18/2023] [Revised: 03/06/2024] [Accepted: 03/13/2024] [Indexed: 03/23/2024]
Abstract
Intestinal disorders can affect pigs of any age, especially when animals are young and more susceptible to infections and environmental stressors. For instance, pathogenic E. coli can alter intestinal functions, thus leading to altered nutrient adsorption by interacting with local cells through lipopolysaccharide (LPS). Among several compounds studied to counteract the negative effects on the intestine, short-chain fatty acids (SCFA) were demonstrated to exert beneficial effects on gut epithelial cells and resident immune cells. In this study, acetate and propionate were tested for their beneficial effects in a co-culture model of IPEC-J2 and porcine PBMC pre-stimulated with LPS from E. coli 0111:B4 aimed at mimicking the interaction between intestinal cells and immune cells in an inflammatory/activated status. IPEC-J2 viability was partially reduced when co-cultured with activated PBMC and nitric oxide concentration increased. IPEC-J2 up-regulated innate and inflammatory markers, namely BD-1, TLR-4, IL-8, TNF-α, NF-κB, and TGF-β. Acetate and propionate positively modulated the inflammatory condition by sustaining cell viability, reducing the oxidative stress, and down-regulating the expression of inflammatory mediators. TNF-α expression and secretion showed an opposite effect in IPEC-J2 depending on the extent of LPS stimulation of PBMC and TGF-β modulation. Therefore, SCFA proved to mediate a differential effect depending on the degree and duration of inflammation. The expression of the tight junction proteins (TJp) claudin-4 and zonula occludens-1 was up-regulated by LPS while SCFA influenced TJp with a different kinetics depending on PBMC stimulation. The co-culture model of IPEC-J2 and LPS-activated PBMC proved to be feasible to address the modulation of markers related to anti-bacterial immunity and inflammation, and intestinal epithelial barrier integrity, which are involved in the in vivo responsiveness and plasticity to infections.
Collapse
Affiliation(s)
- Melania Andrani
- Department of Veterinary Science, University of Parma, Strada del Taglio 10, 43126 Parma, Italy.
| | - Luca Ferrari
- Department of Veterinary Science, University of Parma, Strada del Taglio 10, 43126 Parma, Italy.
| | - Paolo Borghetti
- Department of Veterinary Science, University of Parma, Strada del Taglio 10, 43126 Parma, Italy.
| | - Valeria Cavalli
- Department of Veterinary Science, University of Parma, Strada del Taglio 10, 43126 Parma, Italy.
| | - Elena De Angelis
- Department of Veterinary Science, University of Parma, Strada del Taglio 10, 43126 Parma, Italy.
| | - Francesca Ravanetti
- Department of Veterinary Science, University of Parma, Strada del Taglio 10, 43126 Parma, Italy.
| | - Eleonora Dall'Olio
- Department of Veterinary Science, University of Parma, Strada del Taglio 10, 43126 Parma, Italy.
| | - Paolo Martelli
- Department of Veterinary Science, University of Parma, Strada del Taglio 10, 43126 Parma, Italy.
| | - Roberta Saleri
- Department of Veterinary Science, University of Parma, Strada del Taglio 10, 43126 Parma, Italy.
| |
Collapse
|
5
|
Schusterova P, Mudronova D, Penazziova KL, Hajduckova V, Csank T. Limosilactobacillus reuteri L26 Biocenol TM and its exopolysaccharide: Their influence on rotavirus-induced immune molecules in enterocyte-like cells. VET MED-CZECH 2024; 69:169-176. [PMID: 38841132 PMCID: PMC11148706 DOI: 10.17221/106/2023-vetmed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 04/16/2024] [Indexed: 06/07/2024] Open
Abstract
This study aimed to evaluate the immunomodulatory effect of the probiotic Limosilactobacillus reuteri L26 BiocenolTM (L26) and its purified exopolysaccharide (EPS) with respect to antiviral innate immune response. In our experiment, we used porcine epithelial IPEC-J2 cells as a model of the intestinal barrier in a homologous infection by porcine Rotavirus A strain OSU6 (RVA). The production of selected molecules of non-specific humoral immunity was evaluated at the mRNA level. The EPS alone significantly increased the level of interferon λ3 (IFN-λ3) mRNA in the non-infected IPEC-J2 cells (P < 0.001). We also tested whether the treatment of IPEC-J2 cells by L26 or EPS influences the replication of RVA by virus titration and real-time PCR. We found that a pre-treatment in combination with subsequent continuous stimulation has no influence on the RVA replication. However, both treatments significantly decreased the RVA-induced production of IFN-λ3 (P < 0.05) and the "SOS" cytokine interleukin 6 (IL-6; P < 0.01), already at the transcription level. In addition, the EPS treatment resulted in significantly increased IL-10 mRNA in the RVA-infected cells. In summary, we assume an immunoregulatory potential of L. reuteri L26 BiocenolTM and its EPS in the local intestinal antiviral immune response.
Collapse
Affiliation(s)
- Petra Schusterova
- Department of Microbiology and Immunology, University of Veterinary Medicine and Pharmacy in Košice, Košice, Slovak Republic
| | - Dagmar Mudronova
- Department of Microbiology and Immunology, University of Veterinary Medicine and Pharmacy in Košice, Košice, Slovak Republic
| | - Katarina Loziakova Penazziova
- Department of Microbiology and Immunology, University of Veterinary Medicine and Pharmacy in Košice, Košice, Slovak Republic
| | - Vanda Hajduckova
- Department of Microbiology and Immunology, University of Veterinary Medicine and Pharmacy in Košice, Košice, Slovak Republic
| | - Tomas Csank
- Department of Microbiology and Immunology, University of Veterinary Medicine and Pharmacy in Košice, Košice, Slovak Republic
| |
Collapse
|
6
|
Ferlisi F, De Ciucis CG, Trabalza-Marinucci M, Fruscione F, Mecocci S, Franzoni G, Zinellu S, Galarini R, Razzuoli E, Cappelli K. Olive Mill Waste-Water Extract Enriched in Hydroxytyrosol and Tyrosol Modulates Host-Pathogen Interaction in IPEC-J2 Cells. Animals (Basel) 2024; 14:564. [PMID: 38396532 PMCID: PMC10886184 DOI: 10.3390/ani14040564] [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: 12/21/2023] [Revised: 01/27/2024] [Accepted: 02/05/2024] [Indexed: 02/25/2024] Open
Abstract
The dietary supplementation of olive oil by-products, including olive mill waste-water (OMWW) in animal diets, is a novel application that allows for their re-utilization and recycling and could potentially decrease the use of antibiotics, antimicrobial resistance risk in livestock species, and the occurrence of intestinal diseases. Salmonella serovar typhimurium is one of the most widespread intestinal pathogens in the world, causing enterocolitis in pigs. The aim of this study was to investigate the effect of an OMWW extract enriched in polyphenols (hydroxytyrosol and tyrosol) in the immune response of an intestinal porcine epithelial cell line (IPEC-J2) following S. typhimurium infection. Cells were pre-treated with OMWW-extract polyphenols (OMWW-EP, 0.35 and 1.4 µg) for 24 h and then infected with S. typhimurium for 1 h. We evaluated bacterial invasiveness and assayed IPEC-J2 gene expression with RT-qPCR and cytokine release with an ELISA test. The obtained results showed that OMWW-EP (1.4 µg) significantly reduced S. typhimurium invasiveness; 0.35 µg decreased the IPEC-J2 gene expression of IL1B, MYD88, DEFB1 and DEFB4A, while 1.4 µg down-regulated IL1B and DEFB4A and increased TGFB1. The cytokine content was unchanged in infected cells. This is the first study demonstrating the in vitro immunomodulatory and antimicrobial activity of OMWW extracts enriched in polyphenols, suggesting a protective role of OMWW polyphenols on the pig intestine and their potential application as feed supplements in farm animals such as pigs.
Collapse
Affiliation(s)
- Flavia Ferlisi
- Department of Veterinary Medicine, University of Perugia, 01623 Perugia, Italy; (F.F.); (S.M.); (K.C.)
| | - Chiara Grazia De Ciucis
- National Reference Center of Veterinary and Comparative Oncology (CEROVEC), Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle d’Aosta, 16129 Genova, Italy; (C.G.D.C.); (F.F.); (E.R.)
| | | | - Floriana Fruscione
- National Reference Center of Veterinary and Comparative Oncology (CEROVEC), Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle d’Aosta, 16129 Genova, Italy; (C.G.D.C.); (F.F.); (E.R.)
| | - Samanta Mecocci
- Department of Veterinary Medicine, University of Perugia, 01623 Perugia, Italy; (F.F.); (S.M.); (K.C.)
| | - Giulia Franzoni
- Department of Animal Health, Istituto Zooprofilattico Sperimentale della Sardegna, 07100 Sassari, Italy; (G.F.); (S.Z.)
| | - Susanna Zinellu
- Department of Animal Health, Istituto Zooprofilattico Sperimentale della Sardegna, 07100 Sassari, Italy; (G.F.); (S.Z.)
| | - Roberta Galarini
- Centro Specialistico Sviluppo Metodi Analitici, Istituto Zooprofilattico Sperimentale dell’Umbria e delle Marche “Togo Rosati”, 06126 Perugia, Italy;
| | - Elisabetta Razzuoli
- National Reference Center of Veterinary and Comparative Oncology (CEROVEC), Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle d’Aosta, 16129 Genova, Italy; (C.G.D.C.); (F.F.); (E.R.)
| | - Katia Cappelli
- Department of Veterinary Medicine, University of Perugia, 01623 Perugia, Italy; (F.F.); (S.M.); (K.C.)
| |
Collapse
|
7
|
Sundaram TS, Addis MF, Giromini C, Rebucci R, Pisanu S, Pagnozzi D, Baldi A. Comprehensive proteomic analysis reveals omega-3 fatty acids to counteract endotoxin-stimulated metabolic dysregulation in porcine enterocytes. Sci Rep 2023; 13:21595. [PMID: 38062040 PMCID: PMC10703801 DOI: 10.1038/s41598-023-48018-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 11/21/2023] [Indexed: 12/18/2023] Open
Abstract
Omega-3 polyunsaturated fatty acids (n-3 PUFA), such as the eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), are reported to beneficially affect the intestinal immunity. The biological pathways modulated by n-3 PUFA during an infection, at the level of intestinal epithelial barrier remain elusive. To address this gap, we investigated the proteomic changes induced by n-3 PUFA in porcine enterocyte cell line (IPEC-J2), in the presence and absence of lipopolysaccharide (LPS) stress conditions using shotgun proteomics analysis integrated with RNA-sequencing technology. A total of 33, 85, and 88 differentially abundant proteins (DAPs) were identified in cells exposed to n-3 PUFA (DHA:EPA), LPS, and n-3 PUFA treatment followed by LPS stimulation, respectively. Functional annotation and pathway analysis of DAPs revealed the modulation of central carbon metabolism, including the glycolysis/gluconeogenesis, pentose phosphate pathway, and oxidative phosphorylation processes. Specifically, LPS caused metabolic dysregulation in enterocytes, which was abated upon prior treatment with n-3 PUFA. Besides, n-3 PUFA supplementation facilitated enterocyte development and lipid homeostasis. Altogether, this work for the first time comprehensively described the biological pathways regulated by n-3 PUFA in enterocytes, particularly during endotoxin-stimulated metabolic dysregulation. Additionally, this study may provide nutritional biomarkers in monitoring the intestinal health of human and animals on n-3 PUFA-based diets.
Collapse
Affiliation(s)
- Tamil Selvi Sundaram
- Department of Veterinary Medicine and Animal Sciences, University of Milan, Via Dell'Università 6, 26900, Lodi, Italy.
- University of Veterinary Medicine and Pharmacy in Košice, Komenského 68/73, 04181, Košice, Slovakia.
| | - Maria Filippa Addis
- Department of Veterinary Medicine and Animal Sciences, University of Milan, Via Dell'Università 6, 26900, Lodi, Italy
| | - Carlotta Giromini
- Department of Veterinary Medicine and Animal Sciences, University of Milan, Via Dell'Università 6, 26900, Lodi, Italy
| | - Raffaella Rebucci
- Department of Veterinary Medicine and Animal Sciences, University of Milan, Via Dell'Università 6, 26900, Lodi, Italy
| | - Salvatore Pisanu
- Porto Conte Ricerche S.R.L, S.P. 55 Porto Conte/Capo Caccia, Loc. Tramariglio 15, 07041, Alghero, Italy
| | - Daniela Pagnozzi
- Porto Conte Ricerche S.R.L, S.P. 55 Porto Conte/Capo Caccia, Loc. Tramariglio 15, 07041, Alghero, Italy
| | - Antonella Baldi
- Department of Veterinary Medicine and Animal Sciences, University of Milan, Via Dell'Università 6, 26900, Lodi, Italy
| |
Collapse
|
8
|
He W, Wang J, Han M, Wang L, Li L, Zhang J, Chen S, Guo J, Zhai X, Yang J. Potential Toxicity and Mechanisms of T-2 and HT-2 Individually or in Combination on the Intestinal Barrier Function of Porcine Small Intestinal Epithelial Cells. Toxins (Basel) 2023; 15:682. [PMID: 38133186 PMCID: PMC10748202 DOI: 10.3390/toxins15120682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 11/21/2023] [Accepted: 11/28/2023] [Indexed: 12/23/2023] Open
Abstract
Under natural conditions, T-2 toxin can be easily metabolized to HT-2 toxin by deacetylation, and T-2 and HT-2 are usually co-contaminated in grain and feed at a high detected rate. Our previous information indicated that T-2 toxin could injure the function of the intestinal barrier, but the combined toxicity and mechanism of T-2 and HT-2 on the intestinal cells of porcines are still unknown. Therefore, we aimed to explore T-2 and HT-2 individually and combined on cellular viability, cell membrane integrity, the expression of tight junction-related proteins, and the generation of inflammatory factors in porcine intestinal epithelial cells (IPEC-J2). The results showed that T-2 and HT-2, individually or in combination, could induce a decrease in cell viability, an increase in LDH release and IL-1, IL-6, and TNF-α generation, and a decrease in the anti-inflammatory factor IL-10. Based on the analysis of immunofluorescence staining, real-time PCR, and western blotting, the tight junction protein expressions of Claudin-1, Occludin, and ZO-1 were significantly decreased in the T-2 and HT-2 individual or combination treated groups compared with the control. Furthermore, all the parameter changes in the T-2 + HT-2 combination group were much more serious than those in the individual dose groups. These results suggest that T-2 and HT-2, individually and in combination, could induce an intestinal function injury related to an inflammatory response and damage to the intestinal barrier function in porcine intestinal epithelial cells. Additionally, T-2 and HT-2 in combination showed a synergistic toxic effect, which will provide a theoretical basis to assess the risk of T-2 + HT-2 co-contamination in porcine feed.
Collapse
Affiliation(s)
- Weihua He
- Institute of Pet Science and Technology, Jiangsu Agri-Animal Husbandry Vocational College, Taizhou 225300, China; (W.H.); (L.W.); (L.L.); (J.Z.); (S.C.); (J.G.)
| | - Jianhua Wang
- Institute for Agri-Food Standards and Testing Technology, Shanghai Academy of Agricultural Sciences, Shanghai 201403, China; (J.W.); (M.H.)
| | - Mengyi Han
- Institute for Agri-Food Standards and Testing Technology, Shanghai Academy of Agricultural Sciences, Shanghai 201403, China; (J.W.); (M.H.)
| | - Lihua Wang
- Institute of Pet Science and Technology, Jiangsu Agri-Animal Husbandry Vocational College, Taizhou 225300, China; (W.H.); (L.W.); (L.L.); (J.Z.); (S.C.); (J.G.)
| | - Ling Li
- Institute of Pet Science and Technology, Jiangsu Agri-Animal Husbandry Vocational College, Taizhou 225300, China; (W.H.); (L.W.); (L.L.); (J.Z.); (S.C.); (J.G.)
| | - Jiahui Zhang
- Institute of Pet Science and Technology, Jiangsu Agri-Animal Husbandry Vocational College, Taizhou 225300, China; (W.H.); (L.W.); (L.L.); (J.Z.); (S.C.); (J.G.)
| | - Siqi Chen
- Institute of Pet Science and Technology, Jiangsu Agri-Animal Husbandry Vocational College, Taizhou 225300, China; (W.H.); (L.W.); (L.L.); (J.Z.); (S.C.); (J.G.)
| | - Jiayi Guo
- Institute of Pet Science and Technology, Jiangsu Agri-Animal Husbandry Vocational College, Taizhou 225300, China; (W.H.); (L.W.); (L.L.); (J.Z.); (S.C.); (J.G.)
| | - Xiaohu Zhai
- Institute of Pet Science and Technology, Jiangsu Agri-Animal Husbandry Vocational College, Taizhou 225300, China; (W.H.); (L.W.); (L.L.); (J.Z.); (S.C.); (J.G.)
| | - Junhua Yang
- Institute for Agri-Food Standards and Testing Technology, Shanghai Academy of Agricultural Sciences, Shanghai 201403, China; (J.W.); (M.H.)
| |
Collapse
|
9
|
Han C, Xu W, Wang J, Hou X, Zhou S, Song Q, Liu X, Li H. Porcine Circovirus 2 Increases the Frequency of Transforming Growth Factor-β via the C35, S36 and V39 Amino Acids of the ORF4. Viruses 2023; 15:1602. [PMID: 37515288 PMCID: PMC10383414 DOI: 10.3390/v15071602] [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: 06/05/2023] [Revised: 07/12/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023] Open
Abstract
Porcine circovirus 2 (PCV2) is one of the most important endemic swine pathogens, inducing immunosuppression in pigs and predisposing them to secondary bacterial or viral infections. Our previous studies show that PCV2 infection stimulated pig intestinal epithelial cells (IPEC-J2) to produce the secretory transforming growth factor-β (TGF-β), which, in turn, caused CD4+ T cells to differentiate into regulatory T cells (Tregs). This may be one of the key mechanisms by which PCV2 induces immunosuppression. Here, we attempt to identify the viral proteins that affect the TGF-β secretion, as well as the key amino acids that are primarily responsible for this occurrence. The three amino acids C35, S36 and V39 of the ORF4 protein are the key sites at which PCV2 induces a large amount of TGF-β production in IPEC-J2 and influences the frequency of Tregs. This may elucidate the regulatory effect of PCV2 on the Tregs differentiation from the perspective of virus structure and intestinal epithelial cell interaction, laying a theoretical foundation for improving the molecular mechanism of PCV2-induced intestinal mucosal immunosuppression in piglets.
Collapse
Affiliation(s)
- Cheng Han
- College of Animal Science and Technology, Beijing University of Agriculture, No. 7 Beinong Road, Beijing 102206, China
| | - Weicheng Xu
- College of Animal Science and Technology, Beijing University of Agriculture, No. 7 Beinong Road, Beijing 102206, China
| | - Jianfang Wang
- College of Animal Science and Technology, Beijing University of Agriculture, No. 7 Beinong Road, Beijing 102206, China
| | - Xiaolin Hou
- College of Animal Science and Technology, Beijing University of Agriculture, No. 7 Beinong Road, Beijing 102206, China
| | - Shuanghai Zhou
- College of Animal Science and Technology, Beijing University of Agriculture, No. 7 Beinong Road, Beijing 102206, China
| | - Qinye Song
- College of Veterinary Medicine, Hebei Agricultural University, Baoding 071000, China
| | - Xuewei Liu
- College of Animal Science and Technology, Beijing University of Agriculture, No. 7 Beinong Road, Beijing 102206, China
| | - Huanrong Li
- College of Animal Science and Technology, Beijing University of Agriculture, No. 7 Beinong Road, Beijing 102206, China
| |
Collapse
|
10
|
Binder ARD, Mussack V, Kirchner B, Pfaffl MW. Uptake and effects of polystyrene nanoplastics in comparison to non-plastic silica nanoparticles on small intestine cells (IPEC-J2). ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 262:115147. [PMID: 37343485 DOI: 10.1016/j.ecoenv.2023.115147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 06/10/2023] [Accepted: 06/13/2023] [Indexed: 06/23/2023]
Abstract
Nanoplastics smaller than 1 µm accumulate as anthropogenic material in the food chain, but only little is known about their uptake and possible effects on potentially strongly exposed cells of the small intestine. The aim of the study was to observe the uptake of 100 nm polystyrene nanoplastics into a non-tumorigenic small intestine cell culture model (IPEC-J2 cells) and to monitor the effects on cell growth and gene regulation, compared to a 100 nm non-plastic silica nanoparticle reference. The intracellular uptake of both types of nanoparticles was proven via (confocal) fluorescence microscopy and complemented with transmission electron microscopy. Fluorescence microscopy showed a growth phase-dependent uptake of nanoparticles into the cells, hence further experiments included different time points related to epithelial closure, determined via electric cell substrate impedance sensing. No retardations in epithelial closure of cells after treatment with polystyrene nanoparticles could be found. In contrast, epithelial cell closure was partly negatively influenced by silica nanoparticles. An increased production of organic nanoparticles, like extracellular vesicles, was not measurable via nanoparticle tracking analysis. An assessment of messenger RNA by next generation sequencing and subsequent pathway analysis revealed that the TP53 pathway was influenced significantly by the polystyrene nanoparticle treatment. In both treatments, dysregulated mRNAs were highly enriched in the NOTCH signaling pathway compared to the non-particle control.
Collapse
Affiliation(s)
- Anna Ronja Dorothea Binder
- Chair of Animal Physiology and Immunology, TUM School of Life Sciences, Technical University of Munich, Weihenstephaner Berg 3, D-85354 Freising, Germany.
| | - Veronika Mussack
- Chair of Animal Physiology and Immunology, TUM School of Life Sciences, Technical University of Munich, Weihenstephaner Berg 3, D-85354 Freising, Germany
| | - Benedikt Kirchner
- Chair of Animal Physiology and Immunology, TUM School of Life Sciences, Technical University of Munich, Weihenstephaner Berg 3, D-85354 Freising, Germany
| | - Michael W Pfaffl
- Chair of Animal Physiology and Immunology, TUM School of Life Sciences, Technical University of Munich, Weihenstephaner Berg 3, D-85354 Freising, Germany
| |
Collapse
|
11
|
Andrani M, Borghetti P, Ravanetti F, Cavalli V, Ferrari L, De Angelis E, Martelli P, Saleri R. Acetate and propionate effects in response to LPS in a porcine intestinal co-culture model. Porcine Health Manag 2023; 9:23. [PMID: 37221609 DOI: 10.1186/s40813-023-00316-y] [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: 12/24/2022] [Accepted: 04/27/2023] [Indexed: 05/25/2023] Open
Abstract
BACKGROUND The interest in acetate and propionate as short chain fatty acids (SCFA) derives from research on alternative strategies to the utilization of antibiotics in pig farms. SCFA have a protective role on the intestinal epithelial barrier and improve intestinal immunity by regulating the inflammatory and immune response. This regulation is associated with an increase in intestinal barrier integrity, mediated by the enhancement of tight junction protein (TJp) functions, which prevent the passage of pathogens through the paracellular space. The purpose of this study was to evaluate the effect of in vitro supplementation with SCFA (5 mM acetate and 1 mM propionate) on viability, nitric oxide (NO) release (oxidative stress), NF-κB gene expression, and gene and protein expression of major TJp (occludin [OCLN], zonula occludens-1 [ZO-1], and claudin-4 [CLDN4]) in a porcine intestinal epithelial cell (IPEC-J2) and peripheral blood mononuclear cell (PBMC) co-culture model upon LPS stimulation, through which an acute inflammatory state was simulated. RESULTS Firstly, the inflammatory stimulus induced by LPS evaluated in the IPEC-J2 monoculture was characterized by a reduction of viability, gene expression of TJp and OCLN protein synthesis, and an increase of NO release. The response evaluated in the co-culture showed that acetate positively stimulated the viability of both untreated and LPS-stimulated IPEC-J2 and reduced the release of NO in LPS-stimulated cells. Acetate also promoted an increase of gene expression of CLDN4, ZO-1, and OCLN, and protein synthesis of CLDN4, OCLN and ZO-1 in untreated and LPS-stimulated cells. Propionate induced a reduction of NO release in both untreated and LPS-stimulated IPEC-J2. In untreated cells, propionate induced an increase of TJp gene expression and of CLDN4 and OCLN protein synthesis. Contrarily, propionate in LPS-stimulated cells induced an increase of CLDN4 and OCLN gene expression and protein synthesis. PBMC were influenced by acetate and propionate supplementation, in that NF-κB expression was strongly downregulated in LPS-stimulated cells. CONCLUSIONS The present study demonstrates the protective effect of acetate and propionate upon acute inflammation by regulating epithelial tight junction expression and protein synthesis in a co-culture model, which simulates the in vivo interaction between epithelial intestinal cells and local immune cells.
Collapse
Affiliation(s)
- Melania Andrani
- Department of Veterinary Science, University of Parma, Strada del Taglio 10, 43126, Parma, Italy.
| | - Paolo Borghetti
- Department of Veterinary Science, University of Parma, Strada del Taglio 10, 43126, Parma, Italy
| | - Francesca Ravanetti
- Department of Veterinary Science, University of Parma, Strada del Taglio 10, 43126, Parma, Italy
| | - Valeria Cavalli
- Department of Veterinary Science, University of Parma, Strada del Taglio 10, 43126, Parma, Italy
| | - Luca Ferrari
- Department of Veterinary Science, University of Parma, Strada del Taglio 10, 43126, Parma, Italy
| | - Elena De Angelis
- Department of Veterinary Science, University of Parma, Strada del Taglio 10, 43126, Parma, Italy
| | - Paolo Martelli
- Department of Veterinary Science, University of Parma, Strada del Taglio 10, 43126, Parma, Italy
| | - Roberta Saleri
- Department of Veterinary Science, University of Parma, Strada del Taglio 10, 43126, Parma, Italy
| |
Collapse
|
12
|
Qin W, Xie Y, Ren Z, Xu C, Sun MA, Yin Z, Bao W. Integrative ATAC-seq and RNA-seq analyses of IPEC-J2 cells reveals porcine transcription and chromatin accessibility changes associated with Escherichia coli F18ac inhibited by Lactobacillus reuteri. Front Microbiol 2023; 14:1101111. [PMID: 36876070 PMCID: PMC9978113 DOI: 10.3389/fmicb.2023.1101111] [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: 11/20/2022] [Accepted: 01/30/2023] [Indexed: 02/18/2023] Open
Abstract
Escherichia coli is the main cause of postweaning diarrhea in pigs, leading to economic loss. As a probiotic, Lactobacillus reuteri has been used to inhibit E. coli in clinical applications; however, its integrative interactions with hosts remain unclear, especially in pigs. Here, we found that L. reuteri effectively inhibited E. coli F18ac adhering to porcine IPEC-J2 cells, and explored the genome-wide transcription and chromatin accessibility landscapes of IPEC-J2 cells by RNA-seq and ATAC-seq. The results showed that some key signal transduction pathways, such as PI3K-AKT and MAPK signaling pathways, were enriched in the differentially expressed genes (DEGs) between E. coli F18ac treatment with and without L. reuteri groups. However, we found less overlap between RNA-seq and ATAC-seq datasets; we speculated that this might be caused by histones modification through ChIP-qPCR detection. Furthermore, we identified the regulation of the actin cytoskeleton pathway and a number of candidate genes (ARHGEF12, EGFR, and DIAPH3) that might be associated with the inhibition of E. coli F18ac adherence to IPEC-J2 cells by L. reuteri. In conclusion, we provide a valuable dataset that can be used to seek potential porcine molecular markers of E. coli F18ac pathogenesis and L. reuteri antibacterial activity, and to guide the antibacterial application of L. reuteri.
Collapse
Affiliation(s)
- Weiyun Qin
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China.,College of Veterinary Medicine, Institute of Comparative Medicine, Yangzhou University, Yangzhou, China
| | - Yunxiao Xie
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Zhanshi Ren
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Chao Xu
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Ming-An Sun
- College of Veterinary Medicine, Institute of Comparative Medicine, Yangzhou University, Yangzhou, China
| | - Zongjun Yin
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui, China
| | - Wenbin Bao
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| |
Collapse
|
13
|
Mecocci S, De Paolis L, Zoccola R, Fruscione F, De Ciucis CG, Chiaradia E, Moccia V, Tognoloni A, Pascucci L, Zoppi S, Zappulli V, Chillemi G, Goria M, Cappelli K, Razzuoli E. Antimicrobial and Immunomodulatory Potential of Cow Colostrum Extracellular Vesicles (ColosEVs) in an Intestinal In Vitro Model. Biomedicines 2022; 10:3264. [PMID: 36552020 PMCID: PMC9775086 DOI: 10.3390/biomedicines10123264] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 12/01/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
Extracellular Vesicles (EVs) are nano-sized double-lipid-membrane-bound structures, acting mainly as signalling mediators between distant cells and, in particular, modulating the immune response and inflammation of targeted cells. Milk and colostrum contain high amounts of EVs that could be exploited as alternative natural systems in antimicrobial fighting. The aim of this study is to evaluate cow colostrum-derived EVs (colosEVs) for their antimicrobial, anti-inflammatory and immunomodulating effects in vitro to assess their suitability as natural antimicrobial agents as a strategy to cope with the drug resistance problem. ColosEVs were evaluated on a model of neonatal calf diarrhoea caused by Escherichia coli infection, a livestock disease where antibiotic therapy often has poor results. Colostrum from Piedmontese cows was collected within 24 h of calving and colosEVs were immediately isolated. IPEC-J2 cell line was pre-treated with colosEVs for 48 h and then infected with EPEC/NTEC field strains for 2 h. Bacterial adherence and IPEC-J2 gene expression analysis (RT-qPCR) of CXCL8, DEFB1, DEFB4A, TLR4, TLR5, NFKB1, MYD88, CGAS, RIGI and STING were evaluated. The colosEVs pre-treatment significantly reduced the ability of EPEC/NTEC strains to adhere to cell surfaces (p = 0.006), suggesting a role of ColosEVs in modulating host−pathogen interactions. Moreover, our results showed a significant decrease in TLR5 (p < 0.05), CGAS (p < 0.05) and STING (p < 0.01) gene expression in cells that were pre-treated with ColosEVs and then infected, thus highlighting a potential antimicrobial activity of ColosEVs. This is the first preliminarily study investigating ColosEV immunomodulatory and anti-inflammatory effects on an in vitro model of neonatal calf diarrhoea, showing its potential as a therapeutic and prophylactic tool.
Collapse
Affiliation(s)
- Samanta Mecocci
- Department of Veterinary Medicine, University of Perugia, 06123 Perugia, Italy
| | - Livia De Paolis
- National Reference Center of Veterinary and Comparative Oncology (CEROVEC), Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle d’Aosta, Piazza Borgo Pila 39-24, 16129 Genova, Italy
| | - Roberto Zoccola
- S.C. Biotecnologie Applicate alle Produzioni, Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle d’Aosta, via Bologna 148, 10154 Torino, Italy
| | - Floriana Fruscione
- National Reference Center of Veterinary and Comparative Oncology (CEROVEC), Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle d’Aosta, Piazza Borgo Pila 39-24, 16129 Genova, Italy
| | - Chiara Grazia De Ciucis
- National Reference Center of Veterinary and Comparative Oncology (CEROVEC), Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle d’Aosta, Piazza Borgo Pila 39-24, 16129 Genova, Italy
| | | | - Valentina Moccia
- Department of Comparative Biomedicine and Food Science, University of Padova, 35020 Legnaro, Italy
| | - Alessia Tognoloni
- Department of Veterinary Medicine, University of Perugia, 06123 Perugia, Italy
| | - Luisa Pascucci
- Department of Veterinary Medicine, University of Perugia, 06123 Perugia, Italy
| | - Simona Zoppi
- S.C. Diagnostica Generale, Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle d’Aosta, via Bologna 148, 10154 Torino, Italy
| | - Valentina Zappulli
- Department of Comparative Biomedicine and Food Science, University of Padova, 35020 Legnaro, Italy
| | - Giovanni Chillemi
- Department for Innovation in Biological, Agro-Food and Forest Systems (DIBAF), University of Tuscia, 01100 Viterbo, Italy
| | - Maria Goria
- S.C. Biotecnologie Applicate alle Produzioni, Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle d’Aosta, via Bologna 148, 10154 Torino, Italy
| | - Katia Cappelli
- Department of Veterinary Medicine, University of Perugia, 06123 Perugia, Italy
| | - Elisabetta Razzuoli
- National Reference Center of Veterinary and Comparative Oncology (CEROVEC), Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle d’Aosta, Piazza Borgo Pila 39-24, 16129 Genova, Italy
| |
Collapse
|
14
|
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
|
15
|
Mecocci S, De Paolis L, Fruscione F, Pietrucci D, De Ciucis CG, Giudici SD, Franzoni G, Chillemi G, Cappelli K, Razzuoli E. In vitro evaluation of immunomodulatory activities of goat milk Extracellular Vesicles (mEVs) in a model of gut inflammation. Res Vet Sci 2022; 152:546-556. [PMID: 36179548 DOI: 10.1016/j.rvsc.2022.09.021] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 09/20/2022] [Accepted: 09/21/2022] [Indexed: 11/19/2022]
Abstract
Gut represents a major immunological defense barrier with mucosal immune system and intestinal epithelial cells (IECs). In all intestinal diseases, in particular inflammatory bowel disease (IBD), both the absorption and the local immune system are compromised and alternative effective therapies are sought after. Extracellular Vesicles (EVs) have the capability to regulate immune cells within the inflammatory microenvironment, by dampening inflammation and restoring intestinal barrier integrity. Recently, the immune-modulatory role of EVs has also been confirmed for milk EVs (mEVs), notable for their easy production, high sample volumes, cost-effective scalable production and non-toxic and non-immunogenic behavior. In this context, the aim of this study was to evaluate goat mEV anti-inflammatory and immuno-modulating effects on an in vitro model (IPEC-J2) of intestinal inflammation through gene expression evaluation with RT-qPCR and cytokine release dosage with ELISA test. After the establishment of a pro-inflammatory environment due to LPS stimuli, IL6, CXCL8, IL12p35, IL12p40, IFNB, IL18, TLR7 and NOS2 resulted significantly up-regulated in stimulated IPEC-J2 cells compared to those of the basal culture. After 48 h of mEV treatment in inflamed IPEC-J2 a partial restoration of initial conditions was detected, with the IL18 and IL12p40 significant down-regulation, and IL12p35, EBI3, TLR7, BD1 and BD3 up-regulation. IL-18 reduced protein production was also detected in supernatants. Moreover, a decrease of MMP9 and NOS2 together with a strong up-regulation of MUC2 indicated a recovery of cellular homeostasis and, therefore, potential beneficial effects on the intestinal mucosa. Nevertheless, 48 h post-treatment, an increased gene expression and protein release of IL-8 was observed. This paper is one of the firsts to assess the effect of goat mEVs and the first one, in particular, of doing this on an in vitro model of gut inflammation. The obtained results show a potential capability of goat mEVs to modulate inflammation and to play beneficial effects on the intestinal mucosa.
Collapse
Affiliation(s)
- Samanta Mecocci
- Department of Veterinary Medicine, University of Perugia, Via San Costanzo 4, 06123 Perugia, Italy; Sports Horse Research Center (CRCS), University of Perugia, 06123 Perugia, Italy.
| | - Livia De Paolis
- National Reference Center of Veterinary and Comparative Oncology (CEROVEC), Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle d'Aosta, Piazza Borgo Pila 39/24, 16129 Genova, Italy.
| | - Floriana Fruscione
- National Reference Center of Veterinary and Comparative Oncology (CEROVEC), Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle d'Aosta, Piazza Borgo Pila 39/24, 16129 Genova, Italy.
| | - Daniele Pietrucci
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, IBIOM, CNR, 70126 Bari, Italy; Department for Innovation in Biological, Agro-food and Forest systems (DIBAF), University of Tuscia, 01100 Viterbo, Italy
| | - Chiara Grazia De Ciucis
- National Reference Center of Veterinary and Comparative Oncology (CEROVEC), Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle d'Aosta, Piazza Borgo Pila 39/24, 16129 Genova, Italy.
| | - Silvia Dei Giudici
- Department of Animal Health, Istituto Zooprofilattico Sperimentale della Sardegna, 07100 Sassari, Italy.
| | - Giulia Franzoni
- Department of Animal Health, Istituto Zooprofilattico Sperimentale della Sardegna, 07100 Sassari, Italy.
| | - Giovanni Chillemi
- Department for Innovation in Biological, Agro-food and Forest systems (DIBAF), University of Tuscia, 01100 Viterbo, Italy.
| | - Katia Cappelli
- Department of Veterinary Medicine, University of Perugia, Via San Costanzo 4, 06123 Perugia, Italy; Sports Horse Research Center (CRCS), University of Perugia, 06123 Perugia, Italy.
| | - Elisabetta Razzuoli
- National Reference Center of Veterinary and Comparative Oncology (CEROVEC), Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle d'Aosta, Piazza Borgo Pila 39/24, 16129 Genova, Italy.
| |
Collapse
|
16
|
Lu Y, Zhao M, Mo J, Lan G, Liang J. Dietary supplementation ellagic acid on the growth, intestinal immune response, microbiota, and inflammation in weaned piglets. Front Vet Sci 2022; 9:980271. [PMID: 36118358 PMCID: PMC9478910 DOI: 10.3389/fvets.2022.980271] [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: 06/28/2022] [Accepted: 08/12/2022] [Indexed: 11/13/2022] Open
Abstract
Piglets are susceptible to weaning stress, which weakens the barrier and immune function of the intestinal mucosa, causes inflammation, and ultimately affects animal growth and development. Ellagic acid (EA) is a natural polyphenol dilactone with various biological functions. However, The mechanisms underlying the effects of EA on animal health are still poorly known. Herein, we examined whether dietary supplementation with EA has a positive effect on growth performance, intestinal health, immune response, microbiota, or inflammation in weaned piglets. Sixty weaned piglets (age, 30 days) were randomly divided into two groups: the control group (basic diet) and the test group (basic diet + 500 g/t EA). The pigs were fed for 40 days under the same feeding and management conditions, and the growth performance of each individual was measured. At the end of the feeding period, samples were collected from the small intestinal mucosa for further analysis. Using these tissues, the transcriptome sequences and intestinal microbial diversity were analyzed in both groups. An inflammation model using small intestinal mucosal epithelial cells (IPEC-J2) was also constructed. Dietary EA supplementation significantly increased the average daily weight gain (ADG) and reduced diarrhea rate and serum diamine oxidase (DAO) levels of weaned piglets. Transcriptome sequencing results revealed 401 differentially expressed genes in the jejunum mucosal tissue of pigs in the control and test groups. Of these, 163 genes were up-regulated and 238 were down-regulated. The down-regulated genes were significantly enriched in 10 pathways (false discovery rate < 0.05), including seven pathways related to immune response. The results of bacterial 16s rDNA sequencing show that EA affects the composition of the intestinal microbiota in the cecum and rectum, and reveal significant differences in the abundances of Prevotella_9, Lactobacillus delbrueckii, and Lactobacillus reuteri between the test and control groups (P < 0.05). Experiments using the inflammation model showed that certain doses of EA promote the proliferation of IPEC-J2 cells, increase the relative mRNA expression levels of tight junction-related proteins (ZO-1 and Occludin), improve the compactness of the intestine, reduce the expression of inflammatory factors TNF-α and IL-6, and significantly reduce LPS-induced inflammation in IPEC-J2 cells. In conclusion, we found for the first time that dietary supplementation of EA affects the gut immune response and promotes the beneficial gut microbiota in weaned piglets, reduces the occurrence of inflammatory responses, and thereby promotes the growth and intestinal health of piglets.
Collapse
Affiliation(s)
- Yujie Lu
- College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Mingwei Zhao
- Guangxi Guiken Jinmao Animal Husbandry Co., Ltd., Nanning, China
| | - Jiayuan Mo
- College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Ganqiu Lan
- College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Jing Liang
- College of Animal Science and Technology, Guangxi University, Nanning, China
- *Correspondence: Jing Liang
| |
Collapse
|
17
|
Saleri R, Borghetti P, Ravanetti F, Cavalli V, Ferrari L, De Angelis E, Andrani M, Martelli P. Effects of different short-chain fatty acids (SCFA) on gene expression of proteins involved in barrier function in IPEC-J2. Porcine Health Manag 2022; 8:21. [PMID: 35590351 PMCID: PMC9118747 DOI: 10.1186/s40813-022-00264-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 04/28/2022] [Indexed: 11/10/2022] Open
Abstract
Background Gut microbial anaerobic fermentation produces short-chain fatty acids (SCFA), which are important substrates for energy metabolism and anabolic processes in mammals. SCFA can regulate the inflammatory response and increase the intestinal barrier integrity by enhancing the tight junction protein (TJp) functions, which prevent the passage of antigens through the paracellular space. The aim of this study was to evaluate the effect of in vitro supplementation with SCFA (acetate, propionate, butyrate, and lactate) at different concentrations on viability, nitric oxide (NO) release (oxidative stress parameter) in cell culture supernatants, and gene expression of TJp (occludin, zonula occludens-1, and claudin-4) and pro-inflammatory pathway-related mediators (β-defensin 1, TNF-α, and NF-κB) in intestinal porcine epithelial cell line J2 (IPEC-J2). Results The SCFA tested showed significant effects on IPEC-J2, which proved to be dependent on the type and specific concentration of the fatty acid. Acetate stimulated cell viability and NO production in a dose-dependent manner (P < 0.05), and specifically, 5 mM acetate activated the barrier response through claudin-4, and immunity through β-defensin 1 (P < 0.05). The same effect on these parameters was shown by propionate supplementation, especially at 1 mM (P < 0.05). Contrarily, lactate and butyrate showed different effects compared to acetate and propionate, as they did not stimulate an increase of cell viability and regulated barrier integrity through zonula occludens-1 and occludin, especially at 30 mM and 0.5 mM, respectively (P < 0.05). Upon supplementation with SCFA, the increase of NO release at low levels proved not to have detrimental effects on IPEC-J2 proliferation/survival, and in the case of acetate and propionate, such levels were associated with beneficial effects. Furthermore, the results showed that SCFA supplementation induced β-defensin 1 (P < 0.05) that, in turn, may have been involved in the inhibition of TNF-α and NF-κB gene expression (P < 0.05). Conclusions The present study demonstrates that the supplementation with specific SCFA in IPEC-J2 can significantly modulate the process of barrier protection, and that particularly acetate and propionate sustain cell viability, low oxidative stress activity and intestinal barrier function.
Collapse
Affiliation(s)
- Roberta Saleri
- Department of Veterinary Science, University of Parma, Strada del Taglio 10, 43126, Parma, Italy
| | - Paolo Borghetti
- Department of Veterinary Science, University of Parma, Strada del Taglio 10, 43126, Parma, Italy
| | - Francesca Ravanetti
- Department of Veterinary Science, University of Parma, Strada del Taglio 10, 43126, Parma, Italy
| | - Valeria Cavalli
- Department of Veterinary Science, University of Parma, Strada del Taglio 10, 43126, Parma, Italy
| | - Luca Ferrari
- Department of Veterinary Science, University of Parma, Strada del Taglio 10, 43126, Parma, Italy
| | - Elena De Angelis
- Department of Veterinary Science, University of Parma, Strada del Taglio 10, 43126, Parma, Italy
| | - Melania Andrani
- Department of Veterinary Science, University of Parma, Strada del Taglio 10, 43126, Parma, Italy.
| | - Paolo Martelli
- Department of Veterinary Science, University of Parma, Strada del Taglio 10, 43126, Parma, Italy
| |
Collapse
|
18
|
Xue D, Yang P, Yang Y, Wang Y, Wu K, Qi D, Wang S. Deoxynivalenol triggers porcine intestinal tight junction disorder through hijacking SLC5A1 and PGC1α-mediated mitochondrial function. Food Chem Toxicol 2022; 163:112921. [PMID: 35307453 DOI: 10.1016/j.fct.2022.112921] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 02/23/2022] [Accepted: 03/12/2022] [Indexed: 01/27/2023]
Abstract
Deoxynivalenol (DON) is a mycotoxin frequently occurring in human and animal food worldwide, which raises increasing public health concerns. Growing evidence suggests that mitochondria is a pivotal molecular target for DON. However, the contribution of mitochondrial dysfunction to the pathogenesis of DON-induced gut epithelial barrier disruption remains poorly understood. In an animal experiment, piglets exposed to 2.89 mg DON/kg feed for 4 weeks showed altered metabolomic profiling in the serum and compromised transcriptome in the jejunum. DON exposure also impaired mitochondrial structure in the jejunal mucosa, corresponding with dysfunction of the tight junctions. In IPEC-J2 cells, metabolomic and transcriptomic analyses revealed that DON exposure perturbed biological processes occurring in the mitochondria and disordered the expression of genes involved in mitochondrial energy metabolism. Fuel utilization from glucose was affected by DON exposure, as were mitochondrial morphological dynamics leading to increased fragmentation. A marked loss of Na+/glucose cotransporter (SLC5A1) and peroxisome proliferator activated receptor-γ co-activator 1α (PGC1α) was observed in DON-treated cells. Taken together, our data highlight the critical role of impaired mitochondrial energy metabolism and mitochondrial biogenesis in abnormal intestinal tight junction upon DON exposure, and provide a potential mitochondrial target for intestinal mucosal restoration following DON exposure.
Collapse
Affiliation(s)
- Dongfang Xue
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
| | - Ping Yang
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
| | - Yanyu Yang
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
| | - Yanan Wang
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
| | - Kuntan Wu
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
| | - Desheng Qi
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
| | - Shuai Wang
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, 430070, China.
| |
Collapse
|
19
|
Marks H, Grześkowiak Ł, Martinez-Vallespin B, Dietz H, Zentek J. Porcine and Chicken Intestinal Epithelial Cell Models for Screening Phytogenic Feed Additives—Chances and Limitations in Use as Alternatives to Feeding Trials. Microorganisms 2022; 10:microorganisms10030629. [PMID: 35336204 PMCID: PMC8951747 DOI: 10.3390/microorganisms10030629] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/11/2022] [Accepted: 03/12/2022] [Indexed: 02/07/2023] Open
Abstract
Numerous bioactive plant additives have shown various positive effects in pigs and chickens. The demand for feed additives of natural origin has increased rapidly in recent years to support the health of farm animals and thus minimize the need for antibiotics and other drugs. Although only in vivo experiments can fully represent their effect on the organism, the establishment of reliable in vitro methods is becoming increasingly important in the goal of reducing the use of animals in experiments. The use of cell models requires strict control of the experimental conditions so that reliability and reproducibility can be achieved. In particular, the intestinal porcine epithelial cell line IPEC-J2 represents a promising model for the development of new additives. It offers the possibility to investigate antioxidative, antimicrobial, anti- or pro-proliferative and antiviral effects. However, the use of IPEC-J2 is limited due to its purely epithelial origin and some differences in its morphology and functionality compared to the in vivo situation. With regard to chickens, the development of a reliable intestinal epithelial cell model has attracted the attention of researchers in recent years. Although a promising model was presented lately, further studies are needed to enable the standardized use of a chicken cell line for testing phytogenic feed additives. Finally, co-cultivation of the currently available cell lines with other cell lines and the development of organoids will open up further application possibilities. Special emphasis was given to the IPEC-J2 cell model. Therefore, all publications that investigated plant derived compounds in this cell line were considered. The section on chicken cell lines is based on publications describing the development of chicken intestinal epithelial cell models.
Collapse
Affiliation(s)
- Hannah Marks
- Institute of Animal Nutrition, Department of Veterinary Medicine, Freie Universität Berlin, Königin-Luise-Str. 49, 14195 Berlin, Germany; (Ł.G.); (B.M.-V.); (J.Z.)
- Kaesler Research Institute, Kaesler Nutrition GmbH, Fischkai 1, 27572 Bremerhaven, Germany;
- Correspondence:
| | - Łukasz Grześkowiak
- Institute of Animal Nutrition, Department of Veterinary Medicine, Freie Universität Berlin, Königin-Luise-Str. 49, 14195 Berlin, Germany; (Ł.G.); (B.M.-V.); (J.Z.)
| | - Beatriz Martinez-Vallespin
- Institute of Animal Nutrition, Department of Veterinary Medicine, Freie Universität Berlin, Königin-Luise-Str. 49, 14195 Berlin, Germany; (Ł.G.); (B.M.-V.); (J.Z.)
| | - Heiko Dietz
- Kaesler Research Institute, Kaesler Nutrition GmbH, Fischkai 1, 27572 Bremerhaven, Germany;
| | - Jürgen Zentek
- Institute of Animal Nutrition, Department of Veterinary Medicine, Freie Universität Berlin, Königin-Luise-Str. 49, 14195 Berlin, Germany; (Ł.G.); (B.M.-V.); (J.Z.)
| |
Collapse
|
20
|
Herrera Franco VH, Pardo Carrasco SC, Parra Suescún JE. Antimicrobials added to the feed of weaned piglets at two ages improves the molecular expression of intestinal barrier proteins. ANIMAL PRODUCTION SCIENCE 2022. [DOI: 10.1071/an21027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
21
|
Pahumunto N, Dahlen G, Teanpaisan R. Evaluation of Potential Probiotic Properties of Lactobacillus and Bacillus Strains Derived from Various Sources for Their Potential Use in Swine Feeding. Probiotics Antimicrob Proteins 2021; 15:479-490. [PMID: 34665429 DOI: 10.1007/s12602-021-09861-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/10/2021] [Indexed: 10/20/2022]
Abstract
Beneficial effects of probiotics are relevant to the various potential properties of individual strains, and they may also relate to the original sources of the probiotic strains. This study aimed to characterize the potential probiotic properties of the strains originating from various sources for probiotics use in swine feeding. A total of 9 potential probiotic strains, seven lactobacilli and 2 bacilli, were examined for antimicrobial production against swine pathogens, adhesion and anti-adhesion of potential probiotic strains to IPEC-J2 cells, aggregation ability, host defense peptide expression, and hemolytic assay. The results highlight that all strains derived from different sources could exhibit probiotic properties, although different abilities were observed. L. rhamnosus SD11 exhibited the highest inhibitory effect against all pathogens compared to other strains. Bacillus licheniformis KMP-9, B. subtilis KMP-N004, and L. fermentum SD7 gave the highest internalization and that related to high abilities of exclusion, competition, and displacement inhibition to pathogens. Such strains also gave a higher co-aggregation to all pathogens compared to other potential probiotic strains. L. rhamnosus GG, L. fermentum SD7, L. rhamnosus SD4, and B. subtilis KMP-N004 had significantly higher pBD-2 mRNA expression than other strains. None of potential probiotic strains showed hemolytic activity. In conclusion, the strains derived from either humans or animals possessed desirable probiotic properties including inhibition against porcine pathogens, adhesion capacity to porcine enterocytes, anti-adhesion pathogens to porcine enterocytes, and modulated innate immunity. Results indicate that these probiotic strains may be good candidates for use in swine feeding to reduce the risk of infection.
Collapse
Affiliation(s)
- Nuntiya Pahumunto
- Common Oral Diseases and Epidemiology Research Center, Hat Yai, Thailand.,Department of Oral Diagnostic Sciences, Faculty of Dentistry, Prince of Songkla University, Hat Yai, Thailand
| | - Gunnar Dahlen
- Department of Oral Microbiology and Immunology, Institute of Odontology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Rawee Teanpaisan
- Common Oral Diseases and Epidemiology Research Center, Hat Yai, Thailand. .,Department of Oral Diagnostic Sciences, Faculty of Dentistry, Prince of Songkla University, Hat Yai, Thailand.
| |
Collapse
|
22
|
Saleri R, Borghetti P, Ravanetti F, Andrani M, Cavalli V, De Angelis E, Ferrari L, Martelli P. A Co-Culture Model of IPEC-J2 and Swine PBMC to Study the Responsiveness of Intestinal Epithelial Cells: The Regulatory Effect of Arginine Deprivation. Animals (Basel) 2021; 11:ani11092756. [PMID: 34573721 PMCID: PMC8465608 DOI: 10.3390/ani11092756] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 09/15/2021] [Accepted: 09/16/2021] [Indexed: 12/15/2022] Open
Abstract
Simple Summary The interest in amino acids comes from their involvement in research on alternative strategies for the utilization of antibiotics on farms. Among several substances used to replace antibiotics, there is arginine, an essential amino acid in newborns and piglets. This amino acid has a protective role in intestinal immune cells and improves intestinal immunity. The purpose of this research was to define a co-culture model, in which intestinal epithelial cells can communicate with peripheral blood mononuclear cells (PBMC) to deepen the effects of arginine deprivation on intestinal epithelial cells over time. The main finding was that the lack of arginine highly impacts on intestinal and immune cells by way of immuno-regulation mediated by the expression of pro- and anti-inflammatory cytokines. The use of this experimental model could allow us to investigate the impact of and interactions between specific nutrients and the complex intestinal environment and, in addition, to assess feed additives to improve health and animal production. Abstract Arginine is a semi-essential amino acid, supplementation with which induces a reduction of intestinal damage and an improvement of intestinal immunity in weaned piglets, but the mechanism is not yet entirely clear. The aim of this study was to characterise a co-culture model by measuring changes in gene expression over time (24 and 48 h) in intestinal IPEC-J2 cells in the presence of immune cells activated with phytohemagglutinin and, consequently, to assess the effectiveness of arginine deprivation or supplementation in modulating the expression of certain cytokines related to the regulation of intestinal cells’ function. The main results show the crucial role of arginine in the viability/proliferation of intestinal cells evaluated by an MTT assay, and in the positive regulation of the expression of pro-inflammatory (TNF-α, IL-1α, IL-6, IL-8) and anti-inflammatory (TGF-β) cytokines. This experimental model could be important for analysing and clarifying the role of nutritional conditions in intestinal immune cells’ functionality and reactivity in pigs as well as the mechanisms of the intestinal defence system. Among the potential applications of our in vitro model of interaction between IEC and the immune system there is the possibility of studying the effect of feed additives to improve animal health and production.
Collapse
|
23
|
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
|
24
|
Gresse R, Chaucheyras-Durand F, Garrido JJ, Denis S, Jiménez-Marín A, Beaumont M, Van de Wiele T, Forano E, Blanquet-Diot S. Pathogen Challenge and Dietary Shift Alter Microbiota Composition and Activity in a Mucin-Associated in vitro Model of the Piglet Colon (MPigut-IVM) Simulating Weaning Transition. Front Microbiol 2021; 12:703421. [PMID: 34349744 PMCID: PMC8328230 DOI: 10.3389/fmicb.2021.703421] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 06/24/2021] [Indexed: 12/13/2022] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) is the principal pathogen responsible for post-weaning diarrhea in newly weaned piglets. Expansion of ETEC at weaning is thought to be the consequence of various stress factors such as transient anorexia, dietary change or increase in intestinal inflammation and permeability, but the exact mechanisms remain to be elucidated. As the use of animal experiments raise more and more ethical concerns, we used a recently developed in vitro model of piglet colonic microbiome and mucobiome, the MPigut-IVM, to evaluate the effects of a simulated weaning transition and pathogen challenge at weaning. Our data suggested that the tested factors impacted the composition and functionality of the MPigut-IVM microbiota. The simulation of weaning transition led to an increase in relative abundance of the Prevotellaceae family which was further promoted by the presence of the ETEC strain. In contrast, several beneficial families such as Bacteroidiaceae or Ruminococcaceae and gut health related short chain fatty acids like butyrate or acetate were reduced upon simulated weaning. Moreover, the incubation of MPigut-IVM filtrated effluents with porcine intestinal cell cultures showed that ETEC challenge in the in vitro model led to an increased expression of pro-inflammatory genes by the porcine cells. This study provides insights about the etiology of a dysbiotic microbiota in post-weaning piglets.
Collapse
Affiliation(s)
- Raphaële Gresse
- INRAE, UMR 454 MEDIS, Université Clermont Auvergne, Clermont-Ferrand, France.,Lallemand SAS, Blagnac, France
| | | | - Juan J Garrido
- Grupo de Genómica y Mejora Animal, Departamento de Genética, Facultad de Veterinaria, Universidad de Córdoba, Córdoba, Spain
| | - Sylvain Denis
- INRAE, UMR 454 MEDIS, Université Clermont Auvergne, Clermont-Ferrand, France
| | - Angeles Jiménez-Marín
- Grupo de Genómica y Mejora Animal, Departamento de Genética, Facultad de Veterinaria, Universidad de Córdoba, Córdoba, Spain
| | - Martin Beaumont
- GenPhySE, INRAE, ENVT, Université de Toulouse, Castanet-Tolosan, France
| | - Tom Van de Wiele
- Center for Microbial Ecology and Technology, Ghent University, Ghent, Belgium
| | - Evelyne Forano
- INRAE, UMR 454 MEDIS, Université Clermont Auvergne, Clermont-Ferrand, France
| | | |
Collapse
|
25
|
Relationship between serum concentration, functional parameters and cell bioenergetics in IPEC-J2 cell line. Histochem Cell Biol 2021; 156:59-67. [PMID: 33725198 DOI: 10.1007/s00418-021-01981-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/03/2021] [Indexed: 01/24/2023]
Abstract
The foetal bovine serum (FBS) concentration could influence functional parameters of IPEC-J2 cells. IPEC-J2 is a non-transformed continuous epithelial cell line that represents an established in vitro model to study porcine gut inflammation and alterations of intestinal integrity. This cell line also represents a good translational model thanks to the high similitudes between pig and human gastrointestinal tract. With the aim to assess if the FBS-dependent functional variations are linked to the bioenergetic aspects, the addition of 5% and 10% FBS in the IPEC-J2 culture medium were tested. Doubling time and TEER measurement indicated that cells cultured at higher FBS dose grow faster and as a more compact monolayer. 10% FBS increases ATP production and mitochondrial oxidative phosphorylation (OxPhos) and does not affect glycolysis. Both at 5% and 10% FBS ATP production mainly comes from OxPhos and FBS concentration does not affect the cell respiration bioenergetic parameters. Noteworthy, IPEC-J2 treated with 5% and 10% FBS have a metabolic potential since both OxPhos and glycolysis increase by > 100% and < 50%, respectively in comparison with baseline metabolism. Moreover, glucose, fatty acids and glutamine constitute the preferred metabolic fuel for mitochondrial respiration at both FBS conditions tested. Accordingly, the cells flexibility to oxidize these substrates shows that IPEC-J2 mitochondria cannot maintain the basal ATP production without oxidizing all the substrates available irrespective of FBS concentration. To sum up, in IPEC-J2 cells OxPhos increases with the FBS-stimulated functional physiological parameters to fulfil ATP requirements.
Collapse
|
26
|
Expression and Functional Characterization of a Novel Antimicrobial Peptide: Human Beta-Defensin 118. BIOMED RESEARCH INTERNATIONAL 2020; 2020:1395304. [PMID: 33224970 PMCID: PMC7673234 DOI: 10.1155/2020/1395304] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 10/27/2020] [Accepted: 10/28/2020] [Indexed: 01/25/2023]
Abstract
Purpose β-Defensin 118 (DEFB118) is a novel host defense peptide (HDP) identified in humans. To evaluate its potentials for future utilization, the DEFB118 gene was expressed in Escherichia coli (E. coli) and the recombinant protein was fully characterized. Methods The DEFB118 protein was obtained by heterologous expression using E. coli Rosetta (DE3). Antibacterial activity of DEFB118 was determined by using various bacterial strains. IPEC-J cells challenged by E. coli K88 were used to determine its influences on inflammatory responses. Results The E. coli transformants yielded more than 250 μg/mL DEFB118 protein after 4 h induction by 1.0 mM IPTG. The DEFB118 was estimated by SDS-PAGE to be 30 kDa, and MALDI-TOF analysis verified that it is a human β-defensin 118. Importantly, the DEFB118 showed antimicrobial activities against both Gram-negative bacteria (E. coli K88 and E. coli DH5α) and Gram-positive bacteria (S. aureus and B. subtilis), with a minimum inhibitory concentration (MIC) of 4 μg/mL. Hemolytic assays showed that DEFB118 had no detrimental impact on cell viability. Additionally, DEFB118 was found to elevate the viability of IPEC-J2 cells upon E. coli K88 challenge. Moreover, DEFB118 significantly decreased cell apoptosis in the late apoptosis phase and downregulated the expression of inflammatory cytokines such as IL-1β and TNF-α in IPEC-J2 cell exposure to E. coli K88. Conclusions These results suggested a novel function of the mammalian defensins, and the antibacterial and anti-inflammatory properties of DEFB118 may allow it as a potential substitute for conventionally used antibiotics or drugs.
Collapse
|
27
|
Behrouzi A, Ashrafian F, Mazaheri H, Lari A, Nouri M, Riazi Rad F, Hoseini Tavassol Z, Siadat SD. The importance of interaction between MicroRNAs and gut microbiota in several pathways. Microb Pathog 2020; 144:104200. [PMID: 32289465 DOI: 10.1016/j.micpath.2020.104200] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 04/04/2020] [Accepted: 04/06/2020] [Indexed: 12/17/2022]
Abstract
The human gut harbors diverse microbes that play a fundamental role in the well-being of their host. Microbiota disruption affects the immune function, metabolism, and causes several diseases. Therefore, understanding how the microbiome is adjusted, and identifying methods for manipulating it is critical. Studies have found that there is an inverse association between MicroRNAs (miRNAs) abundance and microbe abundance. miRNAs are known to be engaged in post-transcription regulation of cell-autonomous gene expression. Recently, they have gained great attention for their proposed roles in cell-to-cell communication, and as biomarkers for human disease. Here, we review recent studies on the role of miRNAs as a component of outer membrane vesicles (OMVs) in the composition of gut microbiota and their significance in the human situation of health and diseases and discuss their effect on inflammatory responses and dysbiosis. Further, we explain how probiotics exert influence on the expression of miRNAs.
Collapse
Affiliation(s)
- Ava Behrouzi
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran; Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran
| | - Fatemeh Ashrafian
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran; Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran
| | - Hoora Mazaheri
- Department of Molecular Biology, Pasteur Institute of Iran, Tehran, Iran
| | - Arezou Lari
- Systems Biomedicine Unit, Pasteur Institute of Iran, Tehran, Iran
| | - Matineh Nouri
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran
| | - Farhad Riazi Rad
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran
| | - Zahra Hoseini Tavassol
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran; Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran
| | - Seyed Davar Siadat
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran; Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran; Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
28
|
Razzuoli E, Vencia W, Modesto P, Franzoni G, Giudici SD, Parisi E, Ferrari A, Amadori M. Yersinia enterocolitica-specific modulation of innate immune responses in jejunal epithelial cells. Vet Microbiol 2020; 242:108596. [PMID: 32122600 DOI: 10.1016/j.vetmic.2020.108596] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 01/21/2020] [Accepted: 01/23/2020] [Indexed: 12/12/2022]
Abstract
Gut is often subject to infection by different pathogens like Y. enterocolitica. To date, biotypes (BTs) 1A have been considered as non-pathogenic, because they do not express plasmid of virulence pYV; however, BTs 1A strains present other chromosomic virulence genes and recent studies suggest an implication of this microorganism in reactive arthritis. Although many studies highlighted the molecular basis of pathogenesis of Ye infection, scanty data are available about several environmental BTs 1A strains, often isolated in cases of foodborne disease but not included in pathogenicity studies. The aim of our work was to verify the ability of different Ye 1A strains to adhere and penetrate IPEC-J2 cells and to modulate intestinal innate immunity. Our results showed that all strains under study were able to adhere and penetrate enterocytes, causing inflammatory responses. Indeed, adhesion and invasion of enterocytes is an essential step in Ye pathogenesis (Fàbrega and Vila, 2012). Moreover, our data suggest the possible involvement of strains Ye2/O:9 in reactive arthritis, due to their ability (i) to penetrate enterocytes as pathogenic Ye1/O:8 strains do, and (ii) to increase IL-6, IL-8, IL-12 and IL-18 release. Lastly, our results confirm that IPEC-J2 cells are a very good model to evaluate host-pathogen interaction, and indicate IL-8, TNF-α, TLRs1 and 4 as possible markers of the ability of Ye strains to penetrate enterocytes. Moreover, we showed that Ye strains differently affect the host's innate immune responses.
Collapse
Affiliation(s)
- Elisabetta Razzuoli
- Laboratory of Diagnostic, Istituto Zooprofilattico Sperimentale Del Piemonte, Liguria e Valle d'Aosta, Piazza Borgo Pila 39-24, 16129 Genova, Italy
| | - Walter Vencia
- Laboratory of Diagnostic, Istituto Zooprofilattico Sperimentale Del Piemonte, Liguria e Valle d'Aosta, Piazza Borgo Pila 39-24, 16129 Genova, Italy
| | - Paola Modesto
- Laboratory of Diagnostic, Istituto Zooprofilattico Sperimentale Del Piemonte, Liguria e Valle d'Aosta, Piazza Borgo Pila 39-24, 16129 Genova, Italy
| | - Giulia Franzoni
- Laboratory of Virology, Istituto Zooprofilattico Sperimentale Della Sardegna, Via Vienna 2, 07100 Sassari, Italy
| | - Silvia Dei Giudici
- Laboratory of Virology, Istituto Zooprofilattico Sperimentale Della Sardegna, Via Vienna 2, 07100 Sassari, Italy
| | - Erica Parisi
- Laboratory of Diagnostic, Istituto Zooprofilattico Sperimentale Del Piemonte, Liguria e Valle d'Aosta, Piazza Borgo Pila 39-24, 16129 Genova, Italy
| | - Angelo Ferrari
- Laboratory of Diagnostic, Istituto Zooprofilattico Sperimentale Del Piemonte, Liguria e Valle d'Aosta, Piazza Borgo Pila 39-24, 16129 Genova, Italy
| | - Massimo Amadori
- Laboratory of Animal Welfare, Clinical Chemistry and Veterinary Immunology, Istituto Zooprofilattico Sperimentale Della Lombardia e Dell'Emilia-Romagna, via A. Bianchi 9, 25124, Brescia, Italy.
| |
Collapse
|
29
|
Qian S, Gao Z, Cao R, Yang K, Cui Y, Li S, Meng X, He Q, Li Z. Transmissible Gastroenteritis Virus Infection Up-Regulates FcRn Expression via Nucleocapsid Protein and Secretion of TGF-β in Porcine Intestinal Epithelial Cells. Front Microbiol 2020; 10:3085. [PMID: 32038538 PMCID: PMC6990134 DOI: 10.3389/fmicb.2019.03085] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 12/20/2019] [Indexed: 12/23/2022] Open
Abstract
Transmissible gastroenteritis virus (TGEV) is a porcine intestinal coronavirus that causes fatal severe watery diarrhea in piglets. The neonatal Fc receptor (FcRn) is the only IgG transport receptor, its expression on mucosal surfaces is triggered upon viral stimulation, which significantly enhances mucosal immunity. We utilized TGEV as a model pathogen to explore the role of FcRn in resisting viral invasion in overall intestinal mucosal immunity. TGEV induced FcRn expression by activating NF-κB signaling in porcine small intestinal epithelial (IPEC-J2) cells, however, the underlying mechanisms are unclear. First, using small interfering RNAs, we found that TGEV up-regulated FcRn expression via TLR3, TLR9 and RIG-I. Moreover, TGEV induced IL-1β, IL-6, IL-8, TGF-β, and TNF-α production. TGF-β-stimulated IPEC-J2 cells highly up-regulated FcRn expression, while treatment with a JNK-specific inhibitor down-regulated the expression. TGEV nucleocapsid (N) protein also enhanced FcRn promoter activity via the NF-κB signaling pathway and its central region (aa 128–252) was essential for FcRn activation. Additionally, N protein-mediated FcRn up-regulation promotes IgG transcytosis. Thus, TGEV N protein and TGF-β up-regulated FcRn expression, further clarifying the molecular mechanism of up-regulation of FcRn expression by TGEV.
Collapse
Affiliation(s)
- Shaoju Qian
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Zitong Gao
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Rui Cao
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Kang Yang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Yijie Cui
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Shaowen Li
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China.,Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture of the People's Republic of China, Wuhan, China
| | - Xianrong Meng
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China.,Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture of the People's Republic of China, Wuhan, China
| | - Qigai He
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China.,Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture of the People's Republic of China, Wuhan, China
| | - Zili Li
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China.,Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture of the People's Republic of China, Wuhan, China
| |
Collapse
|
30
|
Peng C, Sun Z, Wang L, Shu Y, He M, Ding H, Li Y, Wang X, Feng S, Li J, Wu J. Soybean antigen protein induces caspase-3/mitochondrion-regulated apoptosis in IPEC-J2 cells. FOOD AGR IMMUNOL 2019. [DOI: 10.1080/09540105.2019.1702926] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Affiliation(s)
- Chenglu Peng
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, People’s Republic of China
| | - Zhifeng Sun
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, People’s Republic of China
| | - Lei Wang
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, People’s Republic of China
| | - Yingshuang Shu
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, People’s Republic of China
| | - Mengchu He
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, People’s Republic of China
| | - Hongyan Ding
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, People’s Republic of China
| | - Yu Li
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, People’s Republic of China
| | - Xichun Wang
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, People’s Republic of China
| | - Shibin Feng
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, People’s Republic of China
| | - Jinchun Li
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, People’s Republic of China
| | - Jinjie Wu
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, People’s Republic of China
| |
Collapse
|
31
|
Šefcová M, Levkut M, Bobíková K, Karaffová V, Revajová V, Maruščáková IC, Levkutová M, Ševčíková Z, Herich R, Levkut M. Cytokine response after stimulation of culture cells by zinc and probiotic strain. In Vitro Cell Dev Biol Anim 2019; 55:830-837. [PMID: 31520371 DOI: 10.1007/s11626-019-00401-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 08/08/2019] [Indexed: 01/06/2023]
Abstract
Intestinal porcine epithelial cells were used for an in vitro analysis of mRNA expression levels of inflammatory cytokines (IL-8, IL-18) and transcriptional factors (MyD88 and NF-κβ). Cells were exposed to inorganic and organic zinc sources (in two different concentrations-50 μmol/L and 100 μmol/L) alone or combined with Lactobacillus reuteri B6/1, which was also applied individually. The total exposure time was 4 h. Quantitative reverse transcriptase PCR was used to determine expression levels of the aforementioned parameters. In general, upregulation was observed; however, a decrease of some mRNA's abundance was also determined. Differences in expression were analysed statistically using ANOVA and Tukey analyses. High relative expression was shown for IL-8, IL-18 and MyD88 in groups treated with 100 μmol/L of inorganic sources of zinc (ZnSO4) (p < 0.05), while groups treated with the organic form did not exhibit significant changes in expression. Also, 50 μmol/L of either zinc source did not significantly modify the transcriptional profile of the cytokines and transcription factors, showing that even inorganic sources, at lower concentrations, do not elicit a significant inflammatory reaction. In summary, supplementation of organic zinc source (Gly-Zn chelate) ensures that IL-8, IL-18, MyD88 and NF-κβ expression levels are not positively regulated. In contrast, inorganic sources of zinc (ZnSO4) could induce an inflammatory reaction. However, this response could be dampened if L. reuteri B6/1 is administered, showing the helpful aspect of using probiotics to modulate an inflammatory response. Conclusively, the use Gly-Zn chelate appears as an optimal alternative for Zn administration that does not compromise normal intestinal homeostasis.
Collapse
Affiliation(s)
- Miroslava Šefcová
- Department of Pathological Anatomy and Pathological Physiology, University of Veterinary Medicine and Pharmacy, Komenského 73, 04181, Košice, Slovak Republic.
| | - Martin Levkut
- Department of Pathological Anatomy and Pathological Physiology, University of Veterinary Medicine and Pharmacy, Komenského 73, 04181, Košice, Slovak Republic
| | - Katarína Bobíková
- Department of Pathological Anatomy and Pathological Physiology, University of Veterinary Medicine and Pharmacy, Komenského 73, 04181, Košice, Slovak Republic
| | - Viera Karaffová
- Department of Pathological Anatomy and Pathological Physiology, University of Veterinary Medicine and Pharmacy, Komenského 73, 04181, Košice, Slovak Republic
| | - Viera Revajová
- Department of Pathological Anatomy and Pathological Physiology, University of Veterinary Medicine and Pharmacy, Komenského 73, 04181, Košice, Slovak Republic
| | - Ivana Cingeľová Maruščáková
- Department of Microbiology and Immunology, University of Veterinary Medicine and Pharmacy, Komenského 73, 04181, Košice, Slovak Republic
| | - Mária Levkutová
- Department of Epizootiology and Parasitology, University of Veterinary Medicine and Pharmacy, Komenského 73, 04181, Košice, Slovak Republic
| | - Zuzana Ševčíková
- Department of Pathological Anatomy and Pathological Physiology, University of Veterinary Medicine and Pharmacy, Komenského 73, 04181, Košice, Slovak Republic
| | - Róbert Herich
- Department of Pathological Anatomy and Pathological Physiology, University of Veterinary Medicine and Pharmacy, Komenského 73, 04181, Košice, Slovak Republic
| | - Mikuláš Levkut
- Department of Pathological Anatomy and Pathological Physiology, University of Veterinary Medicine and Pharmacy, Komenského 73, 04181, Košice, Slovak Republic
- Institute of Neuroimmunology, Slovak Academy of Science, Dúbravská cesta 9, 84510, Bratislava, Slovak Republic
| |
Collapse
|
32
|
Peng C, Ding X, Zhu L, He M, Shu Y, Zhang Y, Li Y, Wang X, Feng S, Li J, Wu J. β-Conglycinin-Induced Intestinal Porcine Epithelial Cell Damage via the Nuclear Factor κB/Mitogen-Activated Protein Kinase Signaling Pathway. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:9009-9021. [PMID: 31319030 DOI: 10.1021/acs.jafc.9b02784] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Soybean allergy is a serious health risk to humans and animals; β-conglycinin is the primary antigenic protein in soybean. Intestinal porcine epithelial (IPEC-J2) cells were used as an in vitro physiological model of the intestinal epithelium to study the effects of different concentrations of soybean antigen protein β-conglycinin to identify the involved signaling pathways. The cells were divided into eight groups and either untreated or treated with different concentrations of β-conglycinin, pyrrolidine dithiocarbamate (PDTC), Nω-nitro-l-arginine methyl ester hydrochloride (l-NAME), SP600125, and SB202190 either alone or in combination. The cells were incubated with 1, 5, and 10 mg·mL-1 β-conglycinin or 5 mg·mL-1 β-conglycinin and 1 μmol·L-1 nuclear factor κB (NF-κB) inhibitor (PDTC), inducible nitric oxide synthase inhibitor (l-NAME), c-Jun N-terminal kinase (JNK) inhibitor (SP600125), and p38 inhibitor (SB202190) for 24 h, separately; controls were left untreated. The mRNA, protein, and phosphorylation levels of NF-κB, p38, and JNK were higher in the treated groups than in the control group. β-Conglycinin decreased tight junction distribution, destroyed the cytoskeleton of IPEC-J2 cells, and caused cell death. After the addition of the inhibitors, β-conglycinin-induced IPEC-J2 cell damage was significantly reduced. β-Conglycinin caused damage to IPEC-J2 cells via the mitogen-activated protein kinase/NF-κB signaling pathway. The results of this study are crucial for exploring the mechanisms underlying allergic reactions caused by soybean antigen proteins.
Collapse
Affiliation(s)
- Chenglu Peng
- College of Animal Science and Technology , Anhui Agricultural University , 130 West Changjiang Road , Hefei , Anhui 230036 , People's Republic of China
| | - Xuedong Ding
- College of Animal Science and Technology , Anhui Agricultural University , 130 West Changjiang Road , Hefei , Anhui 230036 , People's Republic of China
| | - Lei Zhu
- College of Animal Science and Technology , Anhui Agricultural University , 130 West Changjiang Road , Hefei , Anhui 230036 , People's Republic of China
| | - Mengchu He
- College of Animal Science and Technology , Anhui Agricultural University , 130 West Changjiang Road , Hefei , Anhui 230036 , People's Republic of China
| | - Yingshuang Shu
- College of Animal Science and Technology , Anhui Agricultural University , 130 West Changjiang Road , Hefei , Anhui 230036 , People's Republic of China
| | - Yu Zhang
- College of Animal Science and Technology , Anhui Agricultural University , 130 West Changjiang Road , Hefei , Anhui 230036 , People's Republic of China
| | - Yu Li
- College of Animal Science and Technology , Anhui Agricultural University , 130 West Changjiang Road , Hefei , Anhui 230036 , People's Republic of China
| | - Xichun Wang
- College of Animal Science and Technology , Anhui Agricultural University , 130 West Changjiang Road , Hefei , Anhui 230036 , People's Republic of China
| | - Shibin Feng
- College of Animal Science and Technology , Anhui Agricultural University , 130 West Changjiang Road , Hefei , Anhui 230036 , People's Republic of China
| | - Jinchun Li
- College of Animal Science and Technology , Anhui Agricultural University , 130 West Changjiang Road , Hefei , Anhui 230036 , People's Republic of China
| | - Jinjie Wu
- College of Animal Science and Technology , Anhui Agricultural University , 130 West Changjiang Road , Hefei , Anhui 230036 , People's Republic of China
| |
Collapse
|
33
|
Zhao Y, Ran Z, Jiang Q, Hu N, Yu B, Zhu L, Shen L, Zhang S, Chen L, Chen H, Jiang J, Chen D. Vitamin D Alleviates Rotavirus Infection through a Microrna-155-5p Mediated Regulation of the TBK1/IRF3 Signaling Pathway In Vivo and In Vitro. Int J Mol Sci 2019; 20:ijms20143562. [PMID: 31330869 PMCID: PMC6678911 DOI: 10.3390/ijms20143562] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 07/09/2019] [Accepted: 07/16/2019] [Indexed: 12/13/2022] Open
Abstract
(1) Background: Vitamin D (VD) plays a vital role in anti-viral innate immunity. However, the role of VD in anti-rotavirus and its mechanism is still unclear. The present study was performed to investigate whether VD alleviates rotavirus (RV) infection through a microRNA-155-5p (miR-155-5p)-mediated regulation of TANK-binding kinase 1 (TBK1)/interferon regulatory factors 3 (IRF3) signaling pathway in vivo and in vitro. (2) Methods: The efficacy of VD treatment was evaluated in DLY pig and IPEC-J2. Dual-luciferase reporter activity assay was performed to verify the role of miR-155-5p in 1α,25-dihydroxy-VD3 (1,25D3) mediating the regulation of the TBK1/IRF3 signaling pathway. (3) Results: A 5000 IU·kg–1 dietary VD3 supplementation attenuated RV-induced the decrease of the villus height and crypt depth (p < 0.05), and up-regulated TBK1, IRF3, and IFN-β mRNA expressions in the jejunum (p < 0.05). Incubation with 1,25D3 significantly decreased the RV mRNA expression and the RV antigen concentration, and increased the TBK1 mRNA and protein levels, and the phosphoprotein IRF3 (p-IRF3) level (p < 0.05). The expression of miR-155-5p was up-regulated in response to an RV infection in vivo and in vitro (p < 0.05). 1,25D3 significantly repressed the up-regulation of miR-155-5p in vivo and in vitro (p < 0.05). Overexpression of miR-155-5p remarkably suppressed the mRNA and protein levels of TBK1 and p-IRF3 (p < 0.01), while the inhibition of miR-155-5p had an opposite effect. Luciferase activity assays confirmed that miR-155-5p regulated RV replication by directly targeting TBK1, and miR-155-5p suppressed the TBK1 protein level (p < 0.01). (4) Conclusions: These results indicate that miR-155-5p is involved in 1,25D3 mediating the regulation of the TBK1/IRF3 signaling pathway by directly targeting TBK1.
Collapse
Affiliation(s)
- Ye Zhao
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China.
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Ya'an 625014, China.
| | - Zhiming Ran
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Ya'an 625014, China
- Institute of Animal Nutrition, Sichuan Agricultural University, Ya'an 625014, China
| | - Qin Jiang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Ningming Hu
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Ya'an 625014, China
- Institute of Animal Nutrition, Sichuan Agricultural University, Ya'an 625014, China
| | - Bing Yu
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Ya'an 625014, China
- Institute of Animal Nutrition, Sichuan Agricultural University, Ya'an 625014, China
| | - Li Zhu
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Linyuan Shen
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Shunhua Zhang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Lei Chen
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Hong Chen
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Ya'an 625014, China
- Institute of Animal Nutrition, Sichuan Agricultural University, Ya'an 625014, China
| | - Jun Jiang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
- Institute of Animal Nutrition, Sichuan Agricultural University, Ya'an 625014, China
| | - Daiwen Chen
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Ya'an 625014, China.
- Institute of Animal Nutrition, Sichuan Agricultural University, Ya'an 625014, China.
| |
Collapse
|
34
|
Ying C, Hong W, Nianhui Z, Chunlei W, Kehe H, Cuiling P. Nontoxic concentrations of OTA aggravate DON-induced intestinal barrier dysfunction in IPEC-J2 cells via activation of NF-κB signaling pathway. Toxicol Lett 2019; 311:114-124. [PMID: 31026484 DOI: 10.1016/j.toxlet.2019.04.021] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 02/07/2019] [Accepted: 04/18/2019] [Indexed: 01/16/2023]
Abstract
Deoxynivalenol (DON) is well-known enteropathogenic mycotoxin which can alter intestinal barrier functions. Consistently, Ochratoxin A (OTA) ingestion has been found to induce intestinal injuries, including inflammation and diarrhea. However, little is known whether OTA aggravates DON-induced toxicity. This study is designed to explore the effects of OTA on DON-induced intestinal barrier function and involved mechanism. Our results showed either DON or OTA could disrupt intestinal barrier function in a time- and dose-dependent manner, as demonstrated by decreased transepithelial electrical resistance (TEER) and increased paracellular permeability to 4 kDa dextran. However, to eliminate the involvement of cell death, nonlethal concentrations of DON and OTA were used in following experiments. The nontoxic concentration of OTA was observed to aggravate DON-induced intestinal barrier dysfunction, accompanied with tight junction disruption (Claudin-3 and Claudin-4). Moreover, nontoxic concentrations of OTA aggravated DON-induced up-regulation of pro-inflammatory cytokines expression and activated nuclear factor-κB (NF-κB) in IPEC-J2 cells. Adding NF-κB inhibitor (PDTC) alleviated the aggravating effects of nontoxic concentrations of OTA on DON-induced intestinal barrier dysfunction and inflammation. These findings indicate that nontoxic concentrations of OTA promoted DON-induced barrier dysfunction via NF-κB signaling pathway. Our experiment suggests that exposure to nontoxic concentrations of toxins also poses potentially harmful effects.
Collapse
Affiliation(s)
- Chen Ying
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China; Institute of Nutritional and Metabolic Disorders in Domestic Animals and Fowls, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China
| | - Wang Hong
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China; Institute of Nutritional and Metabolic Disorders in Domestic Animals and Fowls, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China
| | - Zhai Nianhui
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China; Institute of Nutritional and Metabolic Disorders in Domestic Animals and Fowls, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China
| | - Wang Chunlei
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China; Institute of Nutritional and Metabolic Disorders in Domestic Animals and Fowls, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China
| | - Huang Kehe
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China; Institute of Nutritional and Metabolic Disorders in Domestic Animals and Fowls, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China.
| | - Pan Cuiling
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China; Institute of Nutritional and Metabolic Disorders in Domestic Animals and Fowls, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China.
| |
Collapse
|
35
|
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
|
36
|
Li Y, Wang J, Li Y, Wu H, Zhao S, Yu Q. Protecting intestinal epithelial cells against deoxynivalenol and E. coli damage by recombinant porcine IL-22. Vet Microbiol 2019; 231:154-159. [PMID: 30955803 PMCID: PMC7172643 DOI: 10.1016/j.vetmic.2019.02.027] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2018] [Revised: 06/01/2018] [Accepted: 02/19/2019] [Indexed: 12/29/2022]
Abstract
Pigs suffer enteritis induced by pathogenic bacteria infection and toxins in the moldy feed, which cause intestinal epithelial damage and diarrhea through the whole breeding cycle. Interleukin-22 (IL-22) plays a critical role in maintaining intestinal mucosal barrier function through repairing intestinal epithelial damage. However, little was known about the effects of IL-22 against apoptosis caused by toxins and infection of intestinal pathogens in the intestinal epithelium, especially in pigs. In this study, we had successfully used prokaryotic expression system to produce recombinant porcine interleukin-22. Meanwhile, purified rIL-22 could activate STAT3 signal pathway and have been demonstrated to be safe to IPEC-J2 cells by increasing E-cadherin expression, without proinflammatory cytokines changes. Furthermore, rIL-22 reversed apoptosis induced by deoxynivalenol (DON) and played a vital part in repairing the intestinal injury. We also found that rIL-22 stimulated epithelial cells to secrete pBD-1 against enterotoxigenic E. coli (ETEC) K88 infection, as well as alleviating apoptosis ratio. This study provided a theoretical basis for curing intestinal inflammation caused by ETEC infection and epithelial apoptosis induced by DON with rIL-22 in pigs.
Collapse
Affiliation(s)
- Yunyun Li
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Weigang 1, Nanjing, Jiangsu, 210095, PR China
| | - Jinquan Wang
- College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi, Xinjiang, 830052, PR China
| | - Yuchen Li
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Weigang 1, Nanjing, Jiangsu, 210095, PR China
| | - Haiqin Wu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Weigang 1, Nanjing, Jiangsu, 210095, PR China
| | - Shiyi Zhao
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Weigang 1, Nanjing, Jiangsu, 210095, PR China
| | - Qinghua Yu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Weigang 1, Nanjing, Jiangsu, 210095, PR China.
| |
Collapse
|
37
|
Zhao Y, Zhang S, Zhang X, Pan L, Bao N, Qin G. Fructooligosaccharide Inhibits the Absorption of β-conglycinin (A Major Soybean Allergen) in IPEC-J2. INTERNATIONAL JOURNAL OF FOOD ENGINEERING 2019; 15. [DOI: 10.1515/ijfe-2018-0116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
Abstract
Abstract
Dissecting the inhibited variation of allergen absorption could contribute to the development of novel therapeutic or preventive treatments for food/feed allergies. This study investigated the effects of fructooligosaccharide (FOS) on the absorption, intracellular accumulation of intact or hydrolysed β-conglycinin in porcine intestinal epithelial cells (IPEC-J2). As demonstrated by ELISA and immunoblotting, β-conglycinin was absorbed in a dose- and time-dependent manner (p < 0.05). Actually, β-conglycinin was easily transported and absorbed after enzymatic hydrolysis. Three peptides (52 kDa, 30 kDa and 25 kDa) were produced during transcellular absorption of intact or hydrolysed β-conglycinin. FOS inhibited the absorption of β-conglycinin, especially the 52 and 30 kDa peptides. The immunoreactive peptides derived from the 52, 35 or 22 kDa peptides were the substrings of the known epitopes determined by mass spectrometry and bioinformatic analyses. These results indicate that FOS can efficiently inhibit the absorption of 52 and 30 kDa peptides derived from β-conglycinin.
Collapse
Affiliation(s)
- Yuan Zhao
- Key Laboratory of Animal Production, Product Quality and Security, Ministry of Education, Jilin Provincial Key Laboratory of Animal nutrition and feed science, College of Animal Science and Technology , Jilin Agricultural University , Changchun 130118 , P.R. China
| | - Shiyao Zhang
- Key Laboratory of Animal Production, Product Quality and Security, Ministry of Education, Jilin Provincial Key Laboratory of Animal nutrition and feed science, College of Animal Science and Technology , Jilin Agricultural University , Changchun 130118 , P.R. China
| | - Xiaodong Zhang
- Institute of Zoonosis, Department of Public Health , Jilin University , Changchun 130062 , P.R. China
| | - Li Pan
- Key Laboratory of Animal Production, Product Quality and Security, Ministry of Education, Jilin Provincial Key Laboratory of Animal nutrition and feed science, College of Animal Science and Technology , Jilin Agricultural University , Changchun 130118 , P.R. China
| | - Nan Bao
- Key Laboratory of Animal Production, Product Quality and Security, Ministry of Education, Jilin Provincial Key Laboratory of Animal nutrition and feed science, College of Animal Science and Technology , Jilin Agricultural University , Changchun 130118 , P.R. China
| | - Guixin Qin
- Key Laboratory of Animal Production, Product Quality and Security, Ministry of Education, Jilin Provincial Key Laboratory of Animal nutrition and feed science, College of Animal Science and Technology , Jilin Agricultural University , Changchun 130118 , P.R. China
| |
Collapse
|
38
|
Giromini C, Cheli F, Rebucci R, Baldi A. Invited review: Dairy proteins and bioactive peptides: Modeling digestion and the intestinal barrier. J Dairy Sci 2018; 102:929-942. [PMID: 30591343 DOI: 10.3168/jds.2018-15163] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 10/11/2018] [Indexed: 12/31/2022]
Abstract
Dairy products are one of the most important sources of biologically active proteins and peptides. The health-promoting functions of these peptides are related to their primary structure, which depends on the parent protein composition. A crucial issue in this field is the demonstration of a cause-effect relationship from the ingested protein form to the bioactive form in vivo. Intervention studies represent the gold standard in nutritional research; however, attention has increasingly been focused on the development of sophisticated in vitro models of digestion to elucidate the mechanism of action of dairy nutrients in a mechanistic way and significantly reduce the number of in vivo trials. On the other hand, the epithelial intestinal barrier is the first gate that actively interacts with digestion metabolites, making the intestinal cells the first target tissue of dairy nutrients and respective metabolites. An evolution of the in vitro digestion approach in the study of dairy proteins and derived bioactive compounds is the setup of combined in vitro digestion and cell culture models taking into consideration the endpoint to measure the target organism (e.g., animal, human) and the key concepts of bioaccessibility, bioavailability, and bioactivity. This review discusses the relevance and challenges of modeling digestion and the intestinal barrier, focusing on the implications for the modeling of dairy protein digestion for bioactivity evaluation.
Collapse
Affiliation(s)
- Carlotta Giromini
- Department of Health, Animal Science and Food Safety, Università degli Studi di Milano, Milan, Italy 20134.
| | - Federica Cheli
- Department of Health, Animal Science and Food Safety, Università degli Studi di Milano, Milan, Italy 20134
| | - Raffaella Rebucci
- Department of Health, Animal Science and Food Safety, Università degli Studi di Milano, Milan, Italy 20134
| | - Antonella Baldi
- Department of Health, Animal Science and Food Safety, Università degli Studi di Milano, Milan, Italy 20134
| |
Collapse
|
39
|
Dou X, Han J, Ma Q, Cheng B, Shan A, Gao N, Yang Y. TLR2/4-mediated NF-κB pathway combined with the histone modification regulates β-defensins and interleukins expression by sodium phenyl butyrate in porcine intestinal epithelial cells. Food Nutr Res 2018; 62:1493. [PMID: 30574051 PMCID: PMC6294838 DOI: 10.29219/fnr.v62.1493] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 10/18/2018] [Accepted: 10/18/2018] [Indexed: 12/13/2022] Open
Abstract
Background Host defense peptides (HDPs) possess direct antibacterial, antineoplastic, and immunomodulatory abilities, playing a vital role in innate immunity. Dietary-regulated HDP holds immense potential as a novel pathway for preventing infection. Objective In this study, we examined the regulation mechanism of HDPs (pEP2C, pBD-1, and pBD-3) and cytokines (IL-8 and IL-18) expression by sodium phenylbutyrate (PBA). Design The effects of PBA on HDP induction and the mechanism involved were studied in porcine intestinal epithelial cell lines (IPEC J2). Results In this study, the results showed that HDPs (pEP2C, pBD-1, and pBD-3) and cytokines (IL-8 and IL-18) expression was increased significantly upon stimulation with PBA in IPEC J2 cells. Furthermore, toll-like receptor 2 (TLR2) and TLR4 were required for the PBA-mediated upregulation of the HDPs. This process occurred and further activated the NF-κB pathway via the phosphorylation of p65 and an IκB α synthesis delay. Meanwhile, histone deacetylase (HDAC) inhibition and an increased phosphorylation of histone H3 on serine S10 also occurred in PBA-induced HDP expression independently with TLR2 and TLR4. Furthermore, p38-MAPK suppressed PBA-induced pEP2C, pBD-1 pBD-3, IL-8, and IL-18 expression, but ERK1/2 failed to abolish the regulation of pBD-3, IL-8, and IL-18. Moreover, epidermal growth factor receptor (EGFR) is involved in PBA-mediated HDP regulation. Conclusions We concluded that PBA induced HDP and cytokine increases but did not cause an excessive pro-inflammatory response, which proceeded through the TLR2 and TLR4-NF-κB pathway and histone modification in IPEC J2 cells.
Collapse
Affiliation(s)
- Xiujing Dou
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, China
| | - Junlan Han
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, China
| | - Qiuyuan Ma
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, China
| | - Baojing Cheng
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, China
| | - Anshan Shan
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, China
| | - Nan Gao
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, China
| | - Yu Yang
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, China
| |
Collapse
|
40
|
Pearce SC, Coia HG, Karl JP, Pantoja-Feliciano IG, Zachos NC, Racicot K. Intestinal in vitro and ex vivo Models to Study Host-Microbiome Interactions and Acute Stressors. Front Physiol 2018; 9:1584. [PMID: 30483150 PMCID: PMC6240795 DOI: 10.3389/fphys.2018.01584] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 10/23/2018] [Indexed: 12/16/2022] Open
Abstract
The gut microbiome is extremely important for maintaining homeostasis with host intestinal epithelial, neuronal, and immune cells and this host-microbe interaction is critical during times of stress or disease. Environmental, nutritional, and cognitive stress are just a few factors known to influence the gut microbiota and are thought to induce microbial dysbiosis. Research on this bidirectional relationship as it pertains to health and disease is extensive and rapidly expanding in both in vivo and in vitro/ex vivo models. However, far less work has been devoted to studying effects of host-microbe interactions on acute stressors and performance, the underlying mechanisms, and the modulatory effects of different stressors on both the host and the microbiome. Additionally, the use of in vitro/ex vivo models to study the gut microbiome and human performance has not been researched extensively nor reviewed. Therefore, this review aims to examine current evidence concerning the current status of in vitro and ex vivo host models, the impact of acute stressors on gut physiology/microbiota as well as potential impacts on human performance and how we can parlay this information for DoD relevance as well as the broader scientific community. Models reviewed include widely utilized intestinal cell models from human and animal models that have been applied in the past for stress or microbiology research as well as ex vivo organ/tissue culture models and new innovative models including organ-on-a-chip and co-culture models.
Collapse
Affiliation(s)
- Sarah C Pearce
- Performance Nutrition Team, Combat Feeding Directorate, Natick Soldier Research, Development and Engineering Center, Natick, MA, United States
| | - Heidi G Coia
- National Research Council, The National Academies of Sciences, Engineering, and Medicine, Washington, DC, United States.,711th Human Performance Wing, Airforce Research Laboratory, Airman Systems Directorate, Human-Centered ISR Division, Molecular Mechanisms Branch, Wright-Patterson Air Force Base, Dayton, OH, United States
| | - J P Karl
- Military Nutrition Division, U.S. Army Research Institute of Environmental Medicine, Natick, MA, United States
| | - Ida G Pantoja-Feliciano
- Soldier Protection and Optimization Directorate, Natick Soldier Research, Development and Engineering Center, Natick, MA, United States
| | - Nicholas C Zachos
- Division of Gastroenterology and Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Kenneth Racicot
- Performance Nutrition Team, Combat Feeding Directorate, Natick Soldier Research, Development and Engineering Center, Natick, MA, United States
| |
Collapse
|
41
|
Fusarium mycotoxins and in vitro species-specific approach with porcine intestinal and brain in vitro barriers: A review. Food Chem Toxicol 2018; 121:666-675. [DOI: 10.1016/j.fct.2018.09.050] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 09/19/2018] [Accepted: 09/21/2018] [Indexed: 01/03/2023]
|
42
|
Chen Z, Yuan Q, Xu G, Chen H, Lei H, Su J. Effects of Quercetin on Proliferation and H₂O₂-Induced Apoptosis of Intestinal Porcine Enterocyte Cells. Molecules 2018; 23:E2012. [PMID: 30103566 PMCID: PMC6222514 DOI: 10.3390/molecules23082012] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 08/05/2018] [Accepted: 08/09/2018] [Indexed: 12/12/2022] Open
Abstract
Weanling stress and toxicosis, which are harmful to the health of pigs' intestines, are associated with oxidative stress. Quercetin (Que) is a polyphenolic compound that shows good anti-cancer, anti-inflammation and anti-oxidation effects. This study aimed to elaborate whether or not Que promotes IPEC-J2 (intestinal porcine enterocyte cells) proliferation and protects IPEC-J2 from oxidative damage. Thus, we examined the effects of Que on proliferation and H₂O₂-induced apoptosis in IPEC-J2. The results showed that Que increased IPEC-J2 viabililty, propelled cells from G1 phase into S phase and down-regulated gene levels of P27 and P21, respectively. Besides, H₂O₂-induced cell damage was alleviated by Que after different exposure times, and Que depressed apoptosis rate, reactive oxygen species (ROS) level and percentage of G1 phase cells and elevated the percentage of cells in G2 phase and S phase and mitochondrial membrane potential (Δψm) after IPEC-J2 exposure to H₂O₂. Meanwhile, Que reduced the value of Bax/Bcl-2 in H₂O₂ exposed cells. In low-degree oxidative damage cells, lipid peroxidation product malondialdehyde (MDA) content and superoxide dismutase (SOD) activity were increased. In turn, Que could reverse the change of MDA content and SOD activity in low-degree damage cells. Nevertheless, catalase (CAT) activity was not changed in IPEC-J2 incubated with Que under low-degree damage conditions. Interestingly, relative expressive levels of the proteins claudin-1 and occludin were not altered under low-degree damage conditions, but Que could improve claudin-1 and occludin levels, slightly. This research indicates that Que can be greatly beneficial for intestinal porcine enterocyte cell proliferation and it protects intestinal porcine enterocyte cells from oxidation-induced apoptosis, and could be used as a potential feed additive for porcine intestinal health against pathogenic factor-induced oxidative damages and apoptosis.
Collapse
Affiliation(s)
- Zhigang Chen
- Department of Basic Veterinary Medicine, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, Hunan, China.
| | - Qiaoling Yuan
- Department of Basic Veterinary Medicine, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, Hunan, China.
| | - Guangren Xu
- Department of Basic Veterinary Medicine, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, Hunan, China.
| | - Huiyu Chen
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, Hunan, China.
| | - Hongyu Lei
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, Hunan, China.
| | - Jianming Su
- Department of Basic Veterinary Medicine, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, Hunan, China.
| |
Collapse
|
43
|
Thøgersen R, Petrat-Melin B, Zamaratskaia G, Grevsen K, Young JF, Rasmussen MK. In vitro effects of rebaudioside A, stevioside and steviol on porcine cytochrome p450 expression and activity. Food Chem 2018; 258:245-253. [DOI: 10.1016/j.foodchem.2018.03.055] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2017] [Revised: 02/23/2018] [Accepted: 03/13/2018] [Indexed: 01/16/2023]
|
44
|
Dou X, Han J, Song W, Dong N, Xu X, Zhang W, Shan A. Sodium butyrate improves porcine host defense peptide expression and relieves the inflammatory response upon Toll-like receptor 2 activation and histone deacetylase inhibition in porcine kidney cells. Oncotarget 2018; 8:26532-26551. [PMID: 28460447 PMCID: PMC5432277 DOI: 10.18632/oncotarget.15714] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2016] [Accepted: 02/13/2017] [Indexed: 12/22/2022] Open
Abstract
Host defense peptides (HDPs) are an important component of the innate immune system and possess direct antimicrobial and immunomodulatory activities. Dietary regulation of HDPs synthesis has emerged as a novel non-antibiotic approach to combat pathogen infection. There are species- and tissue-dependent characteristics of the regulation and mechanism of HDPs. In this study, we investigated whether the histone deacetylase inhibitor (HDACi) sodium butyrate (NaB) could induce HDP expression and the mechanism underlying NaB-regulated HDP expression in PK-15 cells. Our results revealed that NaB augmented HDP expression in PK-15 cells, including porcine β-defensin 3 (pBD3), epididymis protein 2 splicing variant C (pEP2C), pBD128, pBD123, and pBD115, but no inflammatory response occurred. Inhibition of HDAC activity was not sufficient to induce the expression of pBD3 and pEP2C in comparisons of NaB and another HDACi, trichostatin A (TSA). Concomitantly, NF-κB activation was involved in the induction of HDP expression by NaB. MAPK pathway inhibition also prevented pBD3 and pEP2C induction by NaB. Furthermore, NaB could still promote pBD3 and pEP2C expression and inhibit IL-6 production in the presence of the toll-like receptor 2 (TLR2) ligand peptidoglycan. Moreover, TLR2 could be activated by both NaB and peptidoglycan, and blocking TLR2 expression suppressed HDP induction. Finally, we further showed that increased pBD3 could decrease cytokine interleukin-18 (IL-18) and increase porcine claudin 15 (pCLDN15) contents, suggesting an immunoregulatory function of pBD3. In conclusion, this work paves the way for using HDACi-NaB to induce porcine kidney defense peptides while limiting the deleterious risk of an inflammatory response.
Collapse
Affiliation(s)
- Xiujing Dou
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin 150030, P.R. China
| | - Junlan Han
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin 150030, P.R. China
| | - Wentao Song
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin 150030, P.R. China
| | - Na Dong
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin 150030, P.R. China
| | - Xinyao Xu
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin 150030, P.R. China
| | - Wei Zhang
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin 150030, P.R. China
| | - Anshan Shan
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin 150030, P.R. China
| |
Collapse
|
45
|
Razzuoli E, Mignone G, Lazzara F, Vencia W, Ferraris M, Masiello L, Vivaldi B, Ferrari A, Bozzetta E, Amadori M. Impact of cadmium exposure on swine enterocytes. Toxicol Lett 2018; 287:92-99. [PMID: 29421334 DOI: 10.1016/j.toxlet.2018.02.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 01/04/2018] [Accepted: 02/03/2018] [Indexed: 02/07/2023]
Abstract
We tested cadmium (Cd2+) effects on porcine IPEC-J2 cells, which represent an in vitro model of the interaction between intestinal cells and both infectious and non-infectious stressors. Accordingly, we investigated the effects of low (2 μM) to moderate (20 μM) concentrations of Cd2+, in terms of pro-inflammatory gene expression and protein release, as well as of infectivity in a Salmonella typhimurium penetration model. Our data showed a significant (P < .001) increase of intracellular Cd2+ after 3, 6 and 24 h of exposure with respect to levels at 1 h. These data showed the ability of IPEC-J2 to absorb Cd2+ as a function of both time and concentration. Also, the absorption of this heavy metal was related to a significant modulation of important pro-inflammatory messengers. In particular, down-regulation of IL-8 was associated with a significant decrease of Salmonella typhimurium ability to penetrate into IPEC-J2 cells, in agreement with a previous study in which an anti-IL 8 antibody could significantly inhibit Salmonella penetration into the same cells (Razzuoli et al., 2017). This finding demonstrates the ability of Cd2+ to affect the outcome of an important host-pathogen relationship. In conclusion, our study highlighted the ability of an environmental pollutant like Cd2+ to modulate innate immune responses in terms of chemokine release and gene expression, and susceptibility to microbial infections.
Collapse
Affiliation(s)
- E Razzuoli
- Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e valle d'Aosta, piazza BorgoPila 24-39, 16129 Genova, Italy.
| | - G Mignone
- Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e valle d'Aosta, piazza BorgoPila 24-39, 16129 Genova, Italy
| | - F Lazzara
- Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e valle d'Aosta, piazza BorgoPila 24-39, 16129 Genova, Italy
| | - W Vencia
- Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e valle d'Aosta, piazza BorgoPila 24-39, 16129 Genova, Italy
| | - M Ferraris
- Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e valle d'Aosta, piazza BorgoPila 24-39, 16129 Genova, Italy
| | - L Masiello
- Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e valle d'Aosta, piazza BorgoPila 24-39, 16129 Genova, Italy
| | - B Vivaldi
- Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e valle d'Aosta, piazza BorgoPila 24-39, 16129 Genova, Italy
| | - A Ferrari
- Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e valle d'Aosta, piazza BorgoPila 24-39, 16129 Genova, Italy
| | - E Bozzetta
- Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e valle d'Aosta, piazza BorgoPila 24-39, 16129 Genova, Italy
| | - M Amadori
- Istituto Zooprofilattico Sperimentale della Lombardia e dell'Emilia-Romagna, via A. Bianchi 9, 25124 Brescia, Italy
| |
Collapse
|
46
|
Tao X, Liu S, Men X, Xu Z. Over-expression of miR-146b and its regulatory role in intestinal epithelial cell viability, proliferation, and apoptosis in piglets. Biol Direct 2017; 12:27. [PMID: 29178964 PMCID: PMC5702241 DOI: 10.1186/s13062-017-0199-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Accepted: 11/13/2017] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Weaning stress affects the small intestine of piglets. MiR-146b is differentially expressed in suckling and weaned piglets. In this study, we evaluated the effects of miR-146b on cell viability, proliferation, and apoptosis in IPEC-J2 cells. RESULTS Transfection with miR-146b mimics successfully increased miR-146b levels by 1000× (P < 0.001). The over-expression of miR-146b significantly promoted the apoptosis (P < 0.01) of IPEC-J2 cells, with no significant effects on cell viability or proliferation. MiR-146b suppressed the luciferase activity of the miR-TLR4-wt by 57% compared with the negative control, while mutation of the miR-146b binding site significantly blocked the suppressive effect (P < 0.05). Western blot results showed that TLR4 levels decreased in IPEC-J2 cells transfected with miR-146b mimics (P < 0.05). CONCLUSIONS The over-expression of miR-146b promotes IPEC-J2 cell apoptosis. TLR4 is a direct target of miR-146b in IPEC-J2 cells. REVIEWERS This article was reviewed by Eugene Berezikov and Jan B Hoek.
Collapse
Affiliation(s)
- Xin Tao
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, Hangzhou, Zhejiang, 310021, People's Republic of China
| | - Shujie Liu
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, Hangzhou, Zhejiang, 310021, People's Republic of China
| | - Xiaoming Men
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, Hangzhou, Zhejiang, 310021, People's Republic of China
| | - Ziwei Xu
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, Hangzhou, Zhejiang, 310021, People's Republic of China.
| |
Collapse
|
47
|
Zhao Y, Liu D, Zhang S, Pan L, Qin G. Different Damage to the Mechanical Barrier Function of IPEC-J2 Induced by Soybean Allergen β-conglycinin Hydrolyzed Peptides. INTERNATIONAL JOURNAL OF FOOD ENGINEERING 2017. [DOI: 10.1515/ijfe-2016-0370] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
AbstractThree major enzyme-hydrolyzed peptides have been produced after simulative digestionin vitroof soybean β-conglycinin. The intestinal barrier of IPEC-J2 induced by β-conglycinin enzyme-hydrolyzed peptides was evaluated in this study. The increased alkaline phosphatase (AP) activity was actually linearly correlated with the incubation time by the hydrolysate, the purified 52 kD peptide, or the mixture of 25 and 30 kD peptides. The MTT and TEER values declined in dose-dependence (0–2 mg/mL,p\lt 0.05) or in time-dependence (2–24 h,p \lt0.05). After treatment with different hydrolyzed peptides, the tight junction expression of claudin-3, claudin-4, occludin, and ZO-1 were reduced (p\lt 0.05). Finally, it is found out that the maximum damage to the epithelial barrier function was induced by the mixture of 25 and 30 kD peptide, whereas the minimum damage was caused by the 52 kD peptide.
Collapse
|
48
|
Mitochondrial pathway is involved in the protective effects of alpha-ketoglutarate on hydrogen peroxide induced damage to intestinal cells. Oncotarget 2017; 8:74820-74835. [PMID: 29088826 PMCID: PMC5650381 DOI: 10.18632/oncotarget.20426] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 06/19/2017] [Indexed: 12/21/2022] Open
Abstract
Alpha-ketoglutarate, a key intermediate in the Krebs cycle, has been reported to benefit intestinal health. We tested whether alpha-ketoglutarate can protect intestinal cells against hydrogen peroxide induced damage and aimed to reveal the underlying mechanism. Intestinal porcine epithelial cell line J2 were cultured in Dulbecco’s Modified Eagle Medium-High glucose with or without alpha-ketoglutarate and hydrogen peroxide. Cell viability, proliferation, mitochondrial respiration, mitochondrial membrane potential, antioxidant function, apoptosis and mitochondrial-dependent apoptotic pathways were determined. Our experiments demonstrated that, first, exposure to 100μM hydrogen peroxide decreased cell viability, DNA synthesis, mitochondrial respiration and antioxidant function, and increased apoptosis. Second, 2mM alpha-ketoglutarate addition attenuated hydrogen peroxide-induced cell cycle arrest, and improved cell viability, DNA synthesis, mitochondrial respiration and antioxidant function. Third, alpha-ketoglutarate enhanced tricarboxylic acid cycle activity, mitochondrial respiration, and decrease the intracellular content of reactive oxygen species. Finally, alpha-ketoglutarate stabilized the mitochondrial membrane potential, increased the ratio of Bcl-2/Bax, decreased the release of cytochrome c and activation of caspase-3, thereby prevented cell apoptosis. Altogether, we proposed that alpha-ketoglutarate protects intestinal cells against hydrogen peroxide-induced damage partly via mitochondria dependent pathway.
Collapse
|
49
|
Liao P, Liao M, Li L, Tan B, Yin Y. Effect of deoxynivalenol on apoptosis, barrier function, and expression levels of genes involved in nutrient transport, mitochondrial biogenesis and function in IPEC-J2 cells. Toxicol Res (Camb) 2017; 6:866-877. [PMID: 30090549 DOI: 10.1039/c7tx00202e] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 08/15/2017] [Indexed: 12/18/2022] Open
Abstract
This study was conducted to determine the effect of 200 ng mL-1 and 2000 ng mL-1 deoxynivalenol (DON) on apoptosis, barrier function, nutrient transporter gene expression, and free amino acid variation as well as on mitochondrial biogenesis and function-related gene expression in the intestinal porcine epithelial cell line J2 (IPEC-J2) for 6 h, 12 h, and 24 h. Exposure to 200 ng mL-1 DON inhibited the cell viability and promoted cell cycle progression from the G2/M phase to the S phase (P < 0.05). The data showed that the IPEC-J2 cell content of free amino acids, such as valine, methionine, leucine, and phenylalanine, was increased (P < 0.05) after treatment for 6 h; the aspartate, threonine, and lysine contents increased (P < 0.05) after treatment for 12 h; and the aspartate, serine, glycine, alanine, isoleucine, leucine, and lysine contents decreased (P < 0.05) after treatment for 24 h. The expression levels of barrier function genes, including zonula occludens 1 (ZO-1), occludin (OCLN), and claudin 1 (CLDN1), showed a significant reduction (P < 0.05). Moreover, the expression levels of differently regulated nutrient transporter genes, including B0,+ amino acid transporter (B0,+AT) and sodium-glucose transporter 1 (SGLT1) genes, showed a significant decrease (P < 0.05), while the Na+-dependent neutral amino acid transporter 2 (ASCT2) and glucose transporter type 2 (GLUT2) showed a significant increase (P < 0.01). The expression levels of cytokine genes, including IL-8, and IL-1β genes, showed a significant increase (P < 0.05). Furthermore, the expression levels of mitochondrial biogenesis and function-related genes, including mitochondrial transcription factor A (TFAM) and nuclear respiratory factor-1 (NRF), mitochondrial single-strand DNA-binding protein (mt SSB) and mitochondrial polymerase r (mt polr), NADH dehydrogenase subunit 4 (ND4) and cytochrome c oxidase (CcOX) IV, CcOX V and cytochrome c (Cyt c), mammalian silencing information regulator-2α (SIRT-1), glucokinase and citrate synthase (CS), showed a significant increase (P < 0.05). Taken together, the present study indicated that 200 and 2000 ng mL-1 DON could affect proliferation and cell cycle progression from the G2/M phase to the S phase and could mediate the expression levels of differently regulated barrier function, nutrient transport, and mitochondrial biogenesis and function-related genes.
Collapse
Affiliation(s)
- Peng Liao
- Key Laboratory of Agro-ecological Processes in Subtropical Region , Institute of Subtropical Agriculture , Chinese Academy of Sciences , 644# Yuandaer Road , Changsha 410125 , Hunan Province , China . ; ; Tel: +86-731-8461-9703
| | - Meifang Liao
- College of Traditional Chinese Medicine , Hunan University of Chinese Medicine , 300# Xueshi Road , Changsha 410208 , Hunan Province , China
| | - Ling Li
- College of Traditional Chinese Medicine , Hunan University of Chinese Medicine , 300# Xueshi Road , Changsha 410208 , Hunan Province , China
| | - Bie Tan
- Key Laboratory of Agro-ecological Processes in Subtropical Region , Institute of Subtropical Agriculture , Chinese Academy of Sciences , 644# Yuandaer Road , Changsha 410125 , Hunan Province , China . ; ; Tel: +86-731-8461-9703
| | - Yulong Yin
- Key Laboratory of Agro-ecological Processes in Subtropical Region , Institute of Subtropical Agriculture , Chinese Academy of Sciences , 644# Yuandaer Road , Changsha 410125 , Hunan Province , China . ; ; Tel: +86-731-8461-9703
| |
Collapse
|
50
|
Razzuoli E, Amadori M, Lazzara F, Bilato D, Ferraris M, Vito G, Ferrari A. Salmonella serovar-specific interaction with jejunal epithelial cells. Vet Microbiol 2017; 207:219-225. [PMID: 28757027 DOI: 10.1016/j.vetmic.2017.07.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 04/01/2017] [Accepted: 07/03/2017] [Indexed: 01/22/2023]
Abstract
Gut is often a receptacle for many different pathogens in feed and/or the environment, such as Salmonella spp. The current knowledge about pathogenicity of Salmonella is restricted to few serotypes, whereas other important ones like S. Coeln, S. Thompson, S. Veneziana, have not been investigated yet in human and animal models. Therefore, the aim of our work was to verify the ability of widespread environmental Salmonella strains to penetrate and modulate innate immunity in pig intestinal IPEC-J2 cells. Our results outline the different ability of Salmonella strains to modulate innate immunity; the expression of the IFN-β gene was increased by S. Typhimurium, S. Ablogame and S. Diarizonae 2, that also caused an inflammatory response in terms of Interleukin (IL)-1β and/or IL-8 gene espression. In particular, IL-8 gene expression and protein release were significantly modulated by 5 Salmonella strains out of 7. Interestingly, S. Typhimurium, S. Coeln and S. Thompson strains, characterized by a peculiar ability to penetrate into IPEC-J2 cells, up-regulated both IL-8 and TNF-α gene expression. Accordingly, blocking IL-8 was shown to decrease the penetration of S. Typhimurium. On the contrary, S. Diarizonae strain 1, showing lesser invasion of IPEC-J2 cells, down-regulated the p38-MAPK pathway, and it did not induce an inflammatory response. Our results confirm that IPEC-J2 cells are a useful model to evaluate host-gut pathogen interaction and indicate IL-8 and TNF-α as possible predictive markers of invasiveness of Salmonella strains in enterocytes.
Collapse
Affiliation(s)
- Elisabetta Razzuoli
- Laboratory of Diagnostics, Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle d'Aosta, Piazza Borgo Pila 39-24, 16129, Genova, Italy
| | - Massimo Amadori
- Laboratory of Cellular Immunology, Istituto Zooprofilattico Sperimentale della Lombardia e dell'Emilia-Romagna, via A. Bianchi 9, 25124, Brescia, Italy.
| | - Fabrizio Lazzara
- Laboratory of Diagnostics, Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle d'Aosta, Piazza Borgo Pila 39-24, 16129, Genova, Italy
| | - Dania Bilato
- Laboratory of Cellular Immunology, Istituto Zooprofilattico Sperimentale della Lombardia e dell'Emilia-Romagna, via A. Bianchi 9, 25124, Brescia, Italy
| | - Monica Ferraris
- Laboratory of Diagnostics, Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle d'Aosta, Piazza Borgo Pila 39-24, 16129, Genova, Italy
| | - Guendalina Vito
- Laboratory of Diagnostics, Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle d'Aosta, Piazza Borgo Pila 39-24, 16129, Genova, Italy
| | - Angelo Ferrari
- Laboratory of Diagnostics, Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle d'Aosta, Piazza Borgo Pila 39-24, 16129, Genova, Italy
| |
Collapse
|