1
|
Dey C, Sommerfeld IK, Bojarová P, Kodra N, Vrbata D, Zimolová Vlachová M, Křen V, Pich A, Elling L. Color-coded galectin fusion proteins as novel tools in biomaterial science. Biomater Sci 2025; 13:1482-1500. [PMID: 39907577 DOI: 10.1039/d4bm01148a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2025]
Abstract
The inherent carbohydrate-binding specificities of human galectins can serve as recognition elements in both biotechnological and biomedical applications. The combination of the carbohydrate-recognition domain (CRD) of galectins fused to peptides or proteins for purification, immobilization, and imaging enables multifunctional utilization within a single protein. We present here a library of color-coded galectin fusion proteins that incorporate a His6-tag, a fluorescent protein, and a SpyCatcher or SpyTag unit to enable immobilization procedures. These galectin fusion proteins exhibit similar binding properties to the non-fused galectins with micromolar apparent binding affinities. N- and C-terminal fusion partners do not interfere with the SpyCatcher/SpyTag immobilization. By applying SpyCatcher/SpyTag-mediated SC-ST-Gal-3 conjugates, we show the stepwise formation of a three-layer ECM-like structure in vitro. Additionally, we demonstrate the SpyCatcher/SpyTag-mediated immobilization of galectins in microgels, which can serve as a transport platform for localized targeting applications. The proof of concept is provided by the galectin-mediated binding of microgels to colorectal cancer cells.
Collapse
Affiliation(s)
- Carina Dey
- Laboratory for Biomaterials, Institute for Biotechnology and Helmholtz-Institute for Biomedical Engineering, RWTH Aachen University, Pauwelsstraße 20, 52074 Aachen, Germany.
| | - Isabel K Sommerfeld
- DWI - Leibniz-Institute for Interactive Materials, e.V. Forckenbeckstr. 50, 52074 Aachen, Germany
- Functional and Interactive Polymers, Institute of Technical and Macromolecular Chemistry, RWTH Aachen University, Worringerweg 2, 52074 Aachen, Germany
| | - Pavla Bojarová
- Laboratory of Biotransformation, Institute of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, Prague 4, 14200, Czech Republic
- Department of Health Care Disciplines and Population Protection, Faculty of Biomedical Engineering, Czech Technical University in Prague, nám. Sítná 3105, 27201 Kladno, Czech Republic
| | - Nikol Kodra
- Laboratory for Biomaterials, Institute for Biotechnology and Helmholtz-Institute for Biomedical Engineering, RWTH Aachen University, Pauwelsstraße 20, 52074 Aachen, Germany.
| | - David Vrbata
- Laboratory of Biotransformation, Institute of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, Prague 4, 14200, Czech Republic
| | - Miluše Zimolová Vlachová
- Laboratory of Biotransformation, Institute of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, Prague 4, 14200, Czech Republic
| | - Vladimír Křen
- Laboratory of Biotransformation, Institute of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, Prague 4, 14200, Czech Republic
| | - Andrij Pich
- DWI - Leibniz-Institute for Interactive Materials, e.V. Forckenbeckstr. 50, 52074 Aachen, Germany
- Functional and Interactive Polymers, Institute of Technical and Macromolecular Chemistry, RWTH Aachen University, Worringerweg 2, 52074 Aachen, Germany
- Aachen Maastricht Institute for Biobased Materials (AMIBM), Maastricht University, Brightlands Chemelot Campus, Urmonderbaan 22, 6167 RD Geleen, The Netherlands
| | - Lothar Elling
- Laboratory for Biomaterials, Institute for Biotechnology and Helmholtz-Institute for Biomedical Engineering, RWTH Aachen University, Pauwelsstraße 20, 52074 Aachen, Germany.
| |
Collapse
|
2
|
Berkhout MD, Ioannou A, de Ram C, Boeren S, Plugge CM, Belzer C. Mucin-driven ecological interactions in an in vitro synthetic community of human gut microbes. Glycobiology 2024; 34:cwae085. [PMID: 39385462 PMCID: PMC11632381 DOI: 10.1093/glycob/cwae085] [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: 06/14/2024] [Revised: 09/27/2024] [Accepted: 10/07/2024] [Indexed: 10/12/2024] Open
Abstract
Specific human gut microbes inhabit the outer mucus layer of the gastrointestinal tract. Certain residents of this niche can degrade the large and complex mucin glycoproteins that constitute this layer and utilise the degradation products for their metabolism. In turn, this microbial mucin degradation drives specific microbiological ecological interactions in the human gut mucus layer. However, the exact nature of these interactions remains unknown. In this study, we designed and studied an in vitro mucin-degrading synthetic community that included mucin O-glycan degraders and cross-feeding microorganisms by monitoring community composition and dynamics through a combination of 16S rRNA gene amplicon sequencing and qPCR, mucin glycan degradation with PGC-LC-MS/MS, production of mucin-degrading enzymes and other proteins through metaproteomics, and metabolite production with HPLC. We demonstrated that specialist and generalist mucin O-glycan degraders stably co-exist and found evidence for cross-feeding relationships. Cross-feeding on the products of mucin degradation by other gut microbes resulted in butyrate production, hydrogenotrophic acetogenesis, sulfate reduction and methanogenesis. Metaproteomics analysis revealed that mucin glycan degraders Akkermansia muciniphila, Bacteroides spp. and Ruminococcus torques together contributed 92% of the total mucin O-glycan degrading enzyme pool of this community. Furthermore, comparative proteomics showed that in response to cultivation in a community compared to monoculture, mucin glycan degraders increased carbohydrate-active enzymes whereas we also found indications for niche differentiation. These results confirm the complexity of mucin-driven microbiological ecological interactions and the intricate role of carbohydrate-active enzymes in the human gut mucus layer.
Collapse
Affiliation(s)
- Maryse D Berkhout
- Laboratory of Microbiology, Wageningen University and Research, Stippeneng 4, Wageningen 6708 WE, The Netherlands
| | - Athanasia Ioannou
- Laboratory of Microbiology, Wageningen University and Research, Stippeneng 4, Wageningen 6708 WE, The Netherlands
| | - Carol de Ram
- Laboratory of Food Chemistry, Wageningen University and Research, Bornse Weilanden 9, Wageningen 6708 WG, The Netherlands
| | - Sjef Boeren
- Laboratory of Biochemistry, Wageningen University and Research, Stippeneng 4, Wageningen 6708 WE, The Netherlands
| | - Caroline M Plugge
- Laboratory of Microbiology, Wageningen University and Research, Stippeneng 4, Wageningen 6708 WE, The Netherlands
| | - Clara Belzer
- Laboratory of Microbiology, Wageningen University and Research, Stippeneng 4, Wageningen 6708 WE, The Netherlands
| |
Collapse
|
3
|
Ojaimi Loibman S, Quintana-Hayashi MP, Santos L, Lindén SK. Aeromonas salmonicida AI-1 and AI-2 quorum sensing pathways are differentially regulated by rainbow trout mucins and during in vivo colonization. FISH & SHELLFISH IMMUNOLOGY 2024; 153:109862. [PMID: 39209006 DOI: 10.1016/j.fsi.2024.109862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 08/26/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
Aeromonas salmonicida is an opportunistic pathogen with relevance for aquaculture. Fish epithelia are covered by a mucus layer, composed mainly by highly glycosylated mucins, which are the first point of contact between fish and pathogens. Quorum sensing (QS), a bacterial communication mechanism through secreted autoinducer signals that governs gene expression, influences bacterial growth and virulence. The main A. salmonicida autoinducers are mediated by the luxS and asaI genes, corresponding to inter- and intraspecies communication, respectively. The aim of this study was to determine the effect of the mucins that pathogens encounter during colonization of the gill and skin on A. salmonicida QS. We found that expression of A. salmonicida asaI, but not luxS, was increased after culture at 20 °C compared to 10 °C. Rainbow trout gill and skin mucins up-regulated asaI expression 2-fold but down-regulated luxS 10-fold. The downregulation of luxS was reflected by a reduction in autoinducer-2 secretion. Mucins isolated from skin had a stronger inhibitory effect than mucins isolated from gills on both luxS expression and A1-2 secretion, consistent with a higher relative abundance of N-Acetylneuraminic acid on skin mucins than on gill mucins. Reduction of AI-2 production by mucins or luxS-deletion lead to a reduced A. salmonicida auto-aggregation. Furthermore, after colonization of the gill, luxS was down regulated whereas asaI expression was upregulated. Both in vivo and in vitro, the expression of luxS and asaI were thus differentially regulated, frequently in an inverse manner. The strong AI-2 inhibiting effect of the skin mucins is likely part of the mucin-based defense against pathogens.
Collapse
Affiliation(s)
| | | | - Licínia Santos
- Department of Medical Chemistry and Cell Biology, University of Gothenburg, Sweden
| | - Sara K Lindén
- Department of Medical Chemistry and Cell Biology, University of Gothenburg, Sweden.
| |
Collapse
|
4
|
Dierick E, Callens C, Bloch Y, Savvides SN, Hark S, Pelzer S, Ducatelle R, Van Immerseel F, Goossens E. Clostridium perfringens chitinases, key enzymes during early stages of necrotic enteritis in broiler chickens. PLoS Pathog 2024; 20:e1012560. [PMID: 39283899 PMCID: PMC11426533 DOI: 10.1371/journal.ppat.1012560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 09/26/2024] [Accepted: 09/03/2024] [Indexed: 09/27/2024] Open
Abstract
The interaction between bacteria and the intestinal mucus is crucial during the early pathogenesis of many enteric diseases in mammals. A critical step in this process employed by both commensal and pathogenic bacteria focuses on the breakdown of the protective layer presented by the intestinal mucus by mucolytic enzymes. C. perfringens type G, the causative agent of necrotic enteritis in broilers, produces two glycosyl hydrolase family 18 chitinases, ChiA and ChiB, which display distinct substrate preferences. Whereas ChiB preferentially processes linear substrates such as chitin, ChiA prefers larger and more branched substrates, such as carbohydrates presented by the chicken intestinal mucus. Here, we show via crystal structures of ChiA and ChiB in the apo and ligand-bound forms that the two enzymes display structural features that explain their substrate preferences providing a structural blueprint for further interrogation of their function and inhibition. This research focusses on the roles of ChiA and ChiB in bacterial proliferation and mucosal attachment, two processes leading to colonization and invasion of the gut. ChiA and ChiB, either supplemented or produced by the bacteria, led to a significant increase in C. perfringens growth. In addition to nutrient acquisition, the importance of chitinases in bacterial attachment to the mucus layer was shown using an in vitro binding assay of C. perfringens to chicken intestinal mucus. Both an in vivo colonization trial and a necrotic enteritis trial were conducted, demonstrating that a ChiA chitinase mutant strain was less capable to colonize the intestine and was hampered in its disease-causing ability as compared to the wild-type strain. Our findings reveal that the pathogen-specific chitinases produced by C. perfringens type G strains play a fundamental role during colonization, suggesting their potential as vaccine targets.
Collapse
Affiliation(s)
- Evelien Dierick
- Livestock Gut Health Team (LiGHT) Ghent, Department of Pathobiology, Pharmacology and Zoological Medicine, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Chana Callens
- Livestock Gut Health Team (LiGHT) Ghent, Department of Pathobiology, Pharmacology and Zoological Medicine, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Yehudi Bloch
- Unit for Structural Biology, Department of Biochemistry and Microbiology, Ghent University, Ghent, Belgium
- Unit for Structural Biology, VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Current address: European Molecular Biology Laboratory, EMBL Hamburg, c/o DESY, Hamburg, Germany
| | - Savvas N. Savvides
- Unit for Structural Biology, Department of Biochemistry and Microbiology, Ghent University, Ghent, Belgium
- Unit for Structural Biology, VIB-UGent Center for Inflammation Research, Ghent, Belgium
| | - Sarah Hark
- Evonik Operations GmbH, Nutrition & Care, Halle, Westfalen, Germany
| | - Stefan Pelzer
- Evonik Operations GmbH, Nutrition & Care, Halle, Westfalen, Germany
| | - Richard Ducatelle
- Livestock Gut Health Team (LiGHT) Ghent, Department of Pathobiology, Pharmacology and Zoological Medicine, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Filip Van Immerseel
- Livestock Gut Health Team (LiGHT) Ghent, Department of Pathobiology, Pharmacology and Zoological Medicine, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Evy Goossens
- Livestock Gut Health Team (LiGHT) Ghent, Department of Pathobiology, Pharmacology and Zoological Medicine, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| |
Collapse
|
5
|
Gonçalves M, Lopes C, Silva P. Comparative histological description of the intestine in platyfish (Xiphophorus maculatus) and swordtail fish (Xiphophorus helleri). Tissue Cell 2024; 87:102306. [PMID: 38237385 DOI: 10.1016/j.tice.2024.102306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 01/09/2024] [Accepted: 01/10/2024] [Indexed: 03/19/2024]
Abstract
This study aimed to provide a comprehensive analysis of the histological structure of intestinal tissues of platyfish (Xiphophorus maculatus) and swordtail fish (Xiphophorus helleri). Specifically, the objectives were: (1) to compare the structural adaptations of their intestines related to their distinct feeding habits, diet, and digestive strategies; and (2) to explore their potential as animal models for intestinal disease research. Through detailed examination of tissue morphology, cell types, and structural features, this study found that both species lack a stomach, with the intestine directly connected to the esophagus. Additionally, this study proposes a new division of the intestine into anterior and posterior segments based on distinct histological characteristics. The anterior segment may be adapted for temporary food storage and digestion and was characterized by elongated epithelial cells and thin intestinal folds. In contrast, the posterior segment displayed shorter villi and higher concentrations of goblet cells. This study is the first to describe in detail the intestinal morphology of platyfish and swordtail fish. These findings contribute significantly to the understanding of the comparative anatomy and physiology of these fish species, highlighting their potential as valuable models for intestinal biology research.
Collapse
Affiliation(s)
- Marta Gonçalves
- Laboratory of Histology and Embryology, Department of Microscopy, School of Medicine and Biomedical Sciences (ICBAS), University of Porto (U.Porto), Rua Jorge Viterbo Ferreira 228, Porto 4050-313, Portugal
| | - Célia Lopes
- Laboratory of Histology and Embryology, Department of Microscopy, School of Medicine and Biomedical Sciences (ICBAS), University of Porto (U.Porto), Rua Jorge Viterbo Ferreira 228, Porto 4050-313, Portugal; Histomorphology, Physiopathology and Applied Toxicology Team, Interdisciplinary Centre of Marine and Environmental Research (CIIMAR/CIMAR), University of Porto (U.Porto), Terminal de Cruzeiros do Porto de Leixões, Av. General Norton de Matos s/n, Matosinhos 4450-208, Portugal
| | - Paula Silva
- Laboratory of Histology and Embryology, Department of Microscopy, School of Medicine and Biomedical Sciences (ICBAS), University of Porto (U.Porto), Rua Jorge Viterbo Ferreira 228, Porto 4050-313, Portugal; NOVA Institute of Communication (ICNOVA), NOVA School of Social Sciences and Humanities, Universidade NOVA de Lisboa, 1069-061 Lisbon, Portugal.
| |
Collapse
|
6
|
Huang JJ, Feng YM, Zheng SM, Yu CL, Zhou RG, Liu MJ, Bo RN, Yu J, Li JG. Eugenol Possesses Colitis Protective Effects: Impacts on the TLR4/MyD88/NF-[Formula: see text]B Pathway, Intestinal Epithelial Barrier, and Macrophage Polarization. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2024; 52:493-512. [PMID: 38480500 DOI: 10.1142/s0192415x24500216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2024]
Abstract
Eugenol (EU) has been shown to ameliorate experimental colitis due to its anti-oxidant and anti-inflammatory bioactivities. In this study, DSS-induced acute colitis was established and applied to clarify the regulation efficacy of EU on intestinal barrier impairment and macrophage polarization imbalance along with the inflammatory response. Besides, the adjusting effect of EU on macrophages was further investigated in vitro. The results confirmed that EU intervention alleviated DSS-induced colitis through methods such as restraining weight loss and colonic shortening and decreasing DAI scores. Microscopic observation manifested that EU maintained the intestinal barrier integrity in line with the mucus barrier and tight junction protection. Furthermore, EU intervention significantly suppressed the activation of TLR4/MyD88/NF-[Formula: see text]B signaling pathways and pro-inflammatory cytokines gene expressions, while enhancing the expressions of anti-inflammatory cytokines. Simultaneously, WB and FCM analyses of the CD86 and CD206 showed that EU could regulate the DSS-induced macrophage polarization imbalance. Overall, our data further elucidated the mechanism of EU's defensive effect on experimental colitis, which is relevant to the protective efficacy of intestinal barriers, inhibition of oxidative stress and excessive inflammatory response, and reprogramming of macrophage polarization. Hence, this study may facilitate a better understanding of the protective action of the EU against UC.
Collapse
Affiliation(s)
- Jun-Jie Huang
- College of Veterinary Medicine, Yangzhou University, Wenhui East Road 48, Yangzhou 225009, P. R. China
| | - Yue-Min Feng
- College of Veterinary Medicine, Yangzhou University, Wenhui East Road 48, Yangzhou 225009, P. R. China
| | - Shu-Mei Zheng
- College of Veterinary Medicine, Yangzhou University, Wenhui East Road 48, Yangzhou 225009, P. R. China
| | - Cheng-Long Yu
- College of Veterinary Medicine, Yangzhou University, Wenhui East Road 48, Yangzhou 225009, P. R. China
| | - Rui-Gang Zhou
- College of Veterinary Medicine, Yangzhou University, Wenhui East Road 48, Yangzhou 225009, P. R. China
| | - Ming-Jiang Liu
- College of Veterinary Medicine, Yangzhou University, Wenhui East Road 48, Yangzhou 225009, P. R. China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, P. R. China
| | - Ruo-Nan Bo
- College of Veterinary Medicine, Yangzhou University, Wenhui East Road 48, Yangzhou 225009, P. R. China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, P. R. China
| | - Jie Yu
- The Affiliated Suqian First People's Hospital of Nanjing Medical University, Suzhi Road 120, Suqian 223800, P. R. China
| | - Jin-Gui Li
- College of Veterinary Medicine, Yangzhou University, Wenhui East Road 48, Yangzhou 225009, P. R. China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, P. R. China
| |
Collapse
|
7
|
Ibrahim D, I Abdel Rahman MM, M Abd El-Ghany A, A A Hassanen E, A Al-Jabr O, A Abd El-Wahab R, Zayed S, Abd El Khalek Salem M, Nabil El Tahawy S, Youssef W, A Tolba H, E Dawod R, Taha R, H Arisha A, T Y Kishawy A. Chlorella vulgaris extract conjugated magnetic iron nanoparticles in nile tilapia (Oreochromis niloticus): Growth promoting, immunostimulant and antioxidant role and combating against the synergistic infection with Ichthyophthirius multifiliis and Aeromonashydrophila. FISH & SHELLFISH IMMUNOLOGY 2024; 145:109352. [PMID: 38171430 DOI: 10.1016/j.fsi.2023.109352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 12/25/2023] [Accepted: 12/29/2023] [Indexed: 01/05/2024]
Abstract
Nile tilapia reared under intensive conditions was more susceptible for Ichthyophthirius multifilii (I. multifiliis) infection eliciting higher mortality, lower productive rate and further bacterial coinfection with Aeromonas hydrophila (A. hydrophila). The higher potency of magnetic field of iron oxide nanoparticles (NPs) can kill pathogens through inhibiting their viability. Herein, coating of Chlorella vulgaris extract (ChVE) with magnetic iron oxide NPs (Mag iron NPs) can create an external magnetic field that facilitates their release inside the targeted tissues. Thus, the current study is focused on application of new functionalized properties of Mag iron NPs in combination with ChVE and their efficacy to alleviate I. multifiliis and subsequent infection with A. hydrophila in Nile tilapia. Four hundred fingerlings were divided into: control group (with no additives), three groups fed control diet supplemented with ChVE, Mag iron NPs and ChVE@Mag iron NPs for 90 days. At the end of feeding trial fish were challenged with I. multifiliis and at 9 days post challenge was coinfected by A. hydrophila. A remarkable higher growth rate and an improved feed conversion ratio were detected in group fed ChVE@Mag iron-NPs. The maximum expression of antioxidant enzymes in skin and gills tissues (GSH-Px, CAT, and SOD) which came in parallel with higher serum activities of these enzymes was identified in groups received ChVE@Mag iron-NPs. Furthermore, group fed a combination of ChVE and Mag iron-NPs showed a boosted immune response (higher lysozyme, IgM, ACH50, and MPO) prior to challenge with I. multifiliis. In contrast, fish fed ChVE@Mag iron-NPs supplemented diet had lower infection (decreased by 62%) and mortality rates (decreased by 84%), as well as less visible white spots (decreased by 92 % at 12 dpi) on the body surfaces and mucous score. Interestingly, post I. multifiliis the excessive inflammatory response in gill and skin tissues was subsided by feeding on ChVE@Mag iron-NPs as proved by down regulation of IL-1β, TNFα, COX-2 and iNOS and upregulation of IL-10, and IgM, IgT and Muc-2 genes. Notably, group exposed to I. multifiliis-showed higher mortality when exposed to Aeromonas hydrophilia (increased by 43 %) while group fed ChVE@Mag iron-NPs exhibited lower morality (2%). Moreover, the bacterial loads of A. hydrophilia in fish infected by I. multifiliis and fed control diet were higher than those received dietary supplement of ChVE, Mag iron-NPs and the most reduced load was obtained in group fed ChVE@Mag iron-NPs at 7 dpi. In conclusion, ChVE@Mag iron-NPs fed fish had stronger immune barrier and antioxidant functions of skin and gills, and better survival following I. multifiliis and A. hydrophilia infection.
Collapse
Affiliation(s)
- Doaa Ibrahim
- Department of Nutrition and Clinical Nutrition, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44511, Egypt.
| | | | - Amany M Abd El-Ghany
- Department of Parasitology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44511, Egypt
| | - Eman A A Hassanen
- Department of Parasitology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44511, Egypt
| | - Omar A Al-Jabr
- Department of Microbiology, College of Veterinary Medicine, King Faisal University, P.O. Box 400, Al-Ahsa 31982, Saudi Arabia
| | - Reham A Abd El-Wahab
- Biochemistry Department, Animal Health Research Institute (AHRI), Mansoura Branch, Agriculture Research Center (ARC), 246 Dokki, Giza 12618, Egypt
| | - Shimaa Zayed
- Biochemistry Department, Animal Health Research Institute (AHRI), Mansoura Branch, Agriculture Research Center (ARC), 246 Dokki, Giza 12618, Egypt
| | - Mona Abd El Khalek Salem
- Department of Fish Diseases, Animal Health Research Institute (AHRI), Agriculture Research Center, Mansoura, Egypt
| | - Shimaa Nabil El Tahawy
- Department of Clinical Pathology, Zagazig Branch, Animal Health Research Institute, Agriculture Research Center, Zagazig 44516, Egypt
| | - Wessam Youssef
- Biotechnology Department, Animal Health Research Institute (AHRI), 246 Dokki, Giza 12618, Egypt
| | - Heba A Tolba
- Department of Fish Health and Management, Central Laboratory of Aquaculture Research (CLAR), AboHamad, Agriculture Research Center (ARC), Egypt
| | - Rehab E Dawod
- Department of Bacteriology, Animal Health Institute, Damietta Branch, Agriculture Research Center (ARC), Dokki, Giza, Egypt
| | - Rahma Taha
- Department of Zoology, Animal Immunology and Physiology, Faculty of Science, Zagazig University, Zagazig 44511, Egypt
| | - Ahmed H Arisha
- Department of Animal Physiology and Biochemistry, Faculty of Veterinary Medicine, Badr University in Cairo (BUC), Badr City, Cairo, Egypt; Department of Physiology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44511, Egypt
| | - Asmaa T Y Kishawy
- Department of Nutrition and Clinical Nutrition, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44511, Egypt
| |
Collapse
|
8
|
Bachar-Wikstrom E, Dhillon B, Gill Dhillon N, Abbo L, Lindén SK, Wikstrom JD. Mass Spectrometry Analysis of Shark Skin Proteins. Int J Mol Sci 2023; 24:16954. [PMID: 38069276 PMCID: PMC10707392 DOI: 10.3390/ijms242316954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 11/21/2023] [Accepted: 11/24/2023] [Indexed: 12/18/2023] Open
Abstract
The mucus layer covering the skin of fish has several roles, including protection against pathogens and mechanical damage in which proteins play a key role. While proteins in the skin mucus layer of various common bony fish species have been explored, the proteins of shark skin mucus remain unexplored. In this pilot study, we examine the protein composition of the skin mucus in spiny dogfish sharks and chain catsharks through mass spectrometry (NanoLC-MS/MS). Overall, we identified 206 and 72 proteins in spiny dogfish (Squalus acanthias) and chain catsharks (Scyliorhinus retifer), respectively. Categorization showed that the proteins belonged to diverse biological processes and that most proteins were cellular albeit a significant minority were secreted, indicative of mucosal immune roles. The secreted proteins are reviewed in detail with emphasis on their immune potentials. Moreover, STRING protein-protein association network analysis showed that proteins of closely related shark species were more similar as compared to a more distantly related shark and a bony fish, although there were also significant overlaps. This study contributes to the growing field of molecular shark studies and provides a foundation for further research into the functional roles and potential human biomedical implications of shark skin mucus proteins.
Collapse
Affiliation(s)
- Etty Bachar-Wikstrom
- Dermatology and Venereology Division, Department of Medicine (Solna), Karolinska Institutet, 17177 Stockholm, Sweden
- Whitman Center, Marine Biological Laboratory, Woods Hole, MA 02543, USA
| | - Braham Dhillon
- Department of Plant Pathology, Fort Lauderdale Research and Education Center, IFAS, University of Florida, Davie, FL 33314, USA
| | - Navi Gill Dhillon
- Department of Biological Sciences, Nova Southeastern University, Davie, FL 33314, USA
| | - Lisa Abbo
- Whitman Center, Marine Biological Laboratory, Woods Hole, MA 02543, USA
| | - Sara K. Lindén
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, 40530 Gothenburg, Sweden
| | - Jakob D. Wikstrom
- Dermatology and Venereology Division, Department of Medicine (Solna), Karolinska Institutet, 17177 Stockholm, Sweden
- Whitman Center, Marine Biological Laboratory, Woods Hole, MA 02543, USA
- Dermato-Venereology Clinic, Karolinska University Hospital, 17176 Stockholm, Sweden
| |
Collapse
|
9
|
Sanahuja I, Fernandez-Alacid L, Torrecillas S, Ruiz A, Vallejos-Vidal E, Firmino JP, Reyes-Lopez FE, Tort L, Tovar-Ramirez D, Ibarz A, Gisbert E. Dietary Debaryomyces hansenii promotes skin and skin mucus defensive capacities in a marine fish model. Front Immunol 2023; 14:1247199. [PMID: 37711618 PMCID: PMC10499179 DOI: 10.3389/fimmu.2023.1247199] [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: 06/25/2023] [Accepted: 08/09/2023] [Indexed: 09/16/2023] Open
Abstract
The present study explores the effects of two supplementation levels of Debaryomyces hansenii (1.1% and 2.2%) as a probiotic in a reference low fish meal-based diet on the skin mucosal tissue in Sparus aurata. This study includes the evaluation of fish performance coupled with a holistic study of the skin mucosa: i) a transcriptomic study of the skin tissue, and ii) the evaluation of its secreted mucus both in terms of skin mucosal-associated biomarkers and its defensive capacity by means of co-culture analysis with two pathogenic bacteria. Results showed that after 70 days of diet administration, fish fed the diet supplemented with D. hansenii at 1.1% presented increased somatic growth and a better feed conversion ratio, compared to fish fed the control diet. In contrast, fish fed the diet including 2.2% of the probiotic presented intermediate values. Regarding gene regulation, the probiotic administration at 1.1% resulted in 712 differentially expressed genes (DEGs), among which 53.4% and 46.6% were up- and down-regulated, respectively. In particular, D. hansenii modulated some skin biological processes related to immunity and metabolism. Specifically, D. hansenii administration induced a strong modulation of some immune biological-related processes (61 DEGs), mainly involved in B- and T-cell regulatory pathways. Furthermore, dietary D. hansenii promoted the skin barrier function by the upregulation of anchoring junction genes (23 DEGs), which reinforces the physical defense against potential skin damage. In contrast, the skin showed modulated genes related to extracellular exosome and membrane organization (50 DEGs). This modulated functioning is of great interest, particularly in relation to the increased skin mucus defensive capacity observed in the bacterial co-culture in vitro trials, which could be related to the increased modulation and exudation of the innate immune components from the skin cells into the mucus. In summary, the modulation of innate immune parameters coupled with increased skin barrier function and cell trafficking potentiates the skin's physical barrier and mucus defensive capacity, while maintaining the skin mucosa's homeostatic immune and metabolic status. These findings confirmed the advantages of D. hansenii supplementation in low fish meal-based diets, demonstrating the probiotic benefits on cultured marine species.
Collapse
Affiliation(s)
- Ignasi Sanahuja
- Aquaculture Program, Institute of Agrifood Research and Technology (IRTA), La Ràpita, Spain
| | - Laura Fernandez-Alacid
- Department of Cell Biology, Physiology, and Immunology, University of Barcelona, Barcelona, Spain
| | - Silvia Torrecillas
- Aquaculture Program, Institute of Agrifood Research and Technology (IRTA), La Ràpita, Spain
| | - Alberto Ruiz
- Aquaculture Program, Institute of Agrifood Research and Technology (IRTA), La Ràpita, Spain
| | - Eva Vallejos-Vidal
- Núcleo de Investigaciones Aplicadas en Ciencias Veterinarias y Agronómicas, Facultad de Medicina Veterinaria y Agronomía, Universidad de Las Américas, Santiago, Chile
| | - Joana P. Firmino
- Aquaculture Program, Institute of Agrifood Research and Technology (IRTA), La Ràpita, Spain
| | | | - Lluis Tort
- Department of Cell Biology, Physiology, and Immunology, Universitat Autonoma de Barcelona, Barcelona, Spain
| | | | - Antoni Ibarz
- Department of Cell Biology, Physiology, and Immunology, University of Barcelona, Barcelona, Spain
| | - Enric Gisbert
- Aquaculture Program, Institute of Agrifood Research and Technology (IRTA), La Ràpita, Spain
| |
Collapse
|
10
|
Sanahuja I, Ruiz A, Firmino JP, Reyes-López FE, Ortiz-Delgado JB, Vallejos-Vidal E, Tort L, Tovar-Ramírez D, Cerezo IM, Moriñigo MA, Sarasquete C, Gisbert E. Debaryomyces hansenii supplementation in low fish meal diets promotes growth, modulates microbiota and enhances intestinal condition in juvenile marine fish. J Anim Sci Biotechnol 2023; 14:90. [PMID: 37422657 DOI: 10.1186/s40104-023-00895-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 05/11/2023] [Indexed: 07/10/2023] Open
Abstract
BACKGROUND The development of a sustainable business model with social acceptance, makes necessary to develop new strategies to guarantee the growth, health, and well-being of farmed animals. Debaryomyces hansenii is a yeast species that can be used as a probiotic in aquaculture due to its capacity to i) promote cell proliferation and differentiation, ii) have immunostimulatory effects, iii) modulate gut microbiota, and/or iv) enhance the digestive function. To provide inside into the effects of D. hansenii on juveniles of gilthead seabream (Sparus aurata) condition, we integrated the evaluation of the main key performance indicators coupled with the integrative analysis of the intestine condition, through histological and microbiota state, and its transcriptomic profiling. RESULTS After 70 days of a nutritional trial in which a diet with low levels of fishmeal (7%) was supplemented with 1.1% of D. hansenii (17.2 × 105 CFU), an increase of ca. 12% in somatic growth was observed together with an improvement in feed conversion in fish fed a yeast-supplemented diet. In terms of intestinal condition, this probiotic modulated gut microbiota without affecting the intestine cell organization, whereas an increase in the staining intensity of mucins rich in carboxylated and weakly sulphated glycoconjugates coupled with changes in the affinity for certain lectins were noted in goblet cells. Changes in microbiota were characterized by the reduction in abundance of several groups of Proteobacteria, especially those characterized as opportunistic groups. The microarrays-based transcriptomic analysis found 232 differential expressed genes in the anterior-mid intestine of S. aurata, that were mostly related to metabolic, antioxidant, immune, and symbiotic processes. CONCLUSIONS Dietary administration of D. hansenii enhanced somatic growth and improved feed efficiency parameters, results that were coupled to an improvement of intestinal condition as histochemical and transcriptomic tools indicated. This probiotic yeast stimulated host-microbiota interactions without altering the intestinal cell organization nor generating dysbiosis, which demonstrated its safety as a feed additive. At the transcriptomic level, D. hansenii promoted metabolic pathways, mainly protein-related, sphingolipid, and thymidylate pathways, in addition to enhance antioxidant-related intestinal mechanisms, and to regulate sentinel immune processes, potentiating the defensive capacity meanwhile maintaining the homeostatic status of the intestine.
Collapse
Affiliation(s)
- Ignasi Sanahuja
- Aquaculture Program, Institute of Agrifood Research and Technology (IRTA), La Ràpita, 43540, Spain
| | - Alberto Ruiz
- Aquaculture Program, Institute of Agrifood Research and Technology (IRTA), La Ràpita, 43540, Spain
| | - Joana P Firmino
- Aquaculture Program, Institute of Agrifood Research and Technology (IRTA), La Ràpita, 43540, Spain
| | - Felipe E Reyes-López
- Centro de Biotecnología Acuícola, Universidad de Santiago de Chile, Santiago, Chile
| | - Juan B Ortiz-Delgado
- Instituto de Ciencias Marinas de Andalucía (ICMAN-CSIC), Avda. República Saharaui nº 2, Campus Universitario Río San Pedro, Puerto Real, Cádiz, 11510, Spain
| | - Eva Vallejos-Vidal
- Núcleo de Investigaciones Aplicadas en Ciencias Veterinarias y Agronómicas, Facultad de Medicina Veterinaria y Agronomía, Universidad de Las Américas, Santiago, Chile
| | - Lluis Tort
- Department of Cell Biology, Physiology, and Immunology, Universitat Autonoma de Barcelona, Barcelona, Spain
| | | | - Isabel M Cerezo
- Department of Microbiology, Instituto de Biotecnología Y Desarrollo Azul (IBYDA), Faculty of Sciences, University of Malaga, 29010, Malaga, Spain
- SCBI, Bioinformatic Unit, University of Malaga, 29590, Malaga, Spain
| | - Miguel A Moriñigo
- Department of Microbiology, Instituto de Biotecnología Y Desarrollo Azul (IBYDA), Faculty of Sciences, University of Malaga, 29010, Malaga, Spain
| | - Carmen Sarasquete
- Instituto de Ciencias Marinas de Andalucía (ICMAN-CSIC), Avda. República Saharaui nº 2, Campus Universitario Río San Pedro, Puerto Real, Cádiz, 11510, Spain
| | - Enric Gisbert
- Aquaculture Program, Institute of Agrifood Research and Technology (IRTA), La Ràpita, 43540, Spain.
| |
Collapse
|
11
|
Szabó C, Kachungwa Lugata J, Ortega ADSV. Gut Health and Influencing Factors in Pigs. Animals (Basel) 2023; 13:ani13081350. [PMID: 37106913 PMCID: PMC10135089 DOI: 10.3390/ani13081350] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 04/07/2023] [Accepted: 04/11/2023] [Indexed: 04/29/2023] Open
Abstract
The gastrointestinal tract (GIT) is a complex, dynamic, and critical part of the body, which plays an important role in the digestion and absorption of ingested nutrients and excreting waste products of digestion. In addition, GIT also plays a vital role in preventing the entry of harmful substances and potential pathogens into the bloodstream. The gastrointestinal tract hosts a significant number of microbes, which throughout their metabolites, directly interact with the hosts. In modern intensive animal farming, many factors can disrupt GIT functions. As dietary nutrients and biologically active substances play important roles in maintaining homeostasis and eubiosis in the GIT, this review aims to summarize the current status of our knowledge on the most important areas.
Collapse
Affiliation(s)
- Csaba Szabó
- Department of Animal Nutrition and Physiology, Faculty of Agriculture and Food Sciences and Environmental Management, University of Debrecen, Böszörményi Street 138, 4032 Debrecen, Hungary
| | - James Kachungwa Lugata
- Department of Animal Nutrition and Physiology, Faculty of Agriculture and Food Sciences and Environmental Management, University of Debrecen, Böszörményi Street 138, 4032 Debrecen, Hungary
- Doctoral School of Animal Science, Faculty of Agriculture and Food Sciences and Environmental Management, University of Debrecen, Böszörményi Street 138, 4032 Debrecen, Hungary
| | - Arth David Sol Valmoria Ortega
- Department of Animal Nutrition and Physiology, Faculty of Agriculture and Food Sciences and Environmental Management, University of Debrecen, Böszörményi Street 138, 4032 Debrecen, Hungary
- Doctoral School of Animal Science, Faculty of Agriculture and Food Sciences and Environmental Management, University of Debrecen, Böszörményi Street 138, 4032 Debrecen, Hungary
| |
Collapse
|
12
|
Ortiz Y, Heredia N, García S. Boundaries That Prevent or May Lead Animals to be Reservoirs of Escherichia coli O104:H4. J Food Prot 2023; 86:100053. [PMID: 36916560 DOI: 10.1016/j.jfp.2023.100053] [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: 11/08/2022] [Revised: 01/18/2023] [Accepted: 01/21/2023] [Indexed: 02/05/2023]
Abstract
Escherichia coli O104:H4, a hybrid serotype carrying virulence factors from enteroaggregative (EAEC) and Shiga toxin-producing (STEC) pathotypes, is the reported cause of a multicountry outbreak in 2011. Evaluation of potential routes of human contamination revealed that this strain is a foodborne pathogen. In contrast to STEC strains, whose main reservoir is cattle, serotype O104:H4 has not been commonly isolated from animals or related environments, suggesting an inability to naturally colonize the gut in hosts other than humans. However, contrary to this view, this strain has been shown to colonize the intestines of experimental animals in infectious studies. In this minireview, we provide a systematic summary of reports highlighting potential evolutionary changes that could facilitate the colonization of new reservoirs by these bacteria.
Collapse
Affiliation(s)
- Yaraymi Ortiz
- Departamento de Microbiología e Inmunología, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, Ciudad Universitaria, San Nicolás de los Garza, NL 66455, Mexico
| | - Norma Heredia
- Departamento de Microbiología e Inmunología, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, Ciudad Universitaria, San Nicolás de los Garza, NL 66455, Mexico
| | - Santos García
- Departamento de Microbiología e Inmunología, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, Ciudad Universitaria, San Nicolás de los Garza, NL 66455, Mexico.
| |
Collapse
|
13
|
Mortensen JS, Bohr SSR, Harloff-Helleberg S, Hatzakis NS, Saaby L, Nielsen HM. Physical and barrier changes in gastrointestinal mucus induced by the permeation enhancer sodium 8-[(2-hydroxybenzoyl)amino]octanoate (SNAC). J Control Release 2022; 352:163-178. [PMID: 36314534 DOI: 10.1016/j.jconrel.2022.09.034] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 09/09/2022] [Accepted: 09/15/2022] [Indexed: 11/08/2022]
Abstract
Drug delivery systems (DDS) for oral delivery of peptide drugs contain excipients that facilitate and enhance absorption. However, little knowledge exists on how DDS excipients such as permeation enhancers interact with the gastrointestinal mucus barrier. This study aimed to investigate interactions of the permeation enhancer sodium 8-[(2-hydroxybenzoyl)amino]octanoate (SNAC) with ex vivo porcine intestinal mucus (PIM), ex vivo porcine gastric mucus (PGM), as well as with in vitro biosimilar mucus (BM) by profiling their physical and barrier properties upon exposure to SNAC. Bulk mucus permeability studies using the peptides cyclosporine A and vancomycin, ovalbumin as a model protein, as well as fluorescein-isothiocyanate dextrans (FDs) of different molecular weights and different surface charges were conducted in parallel to mucus retention force studies using a texture analyzer, rheological studies, cryo-scanning electron microscopy (cryo-SEM), and single particle tracking of fluorescence-labelled nanoparticles to investigate the effects of the SNAC-mucus interaction. The exposure of SNAC to PIM increased the mucus retention force, storage modulus, viscosity, increased nanoparticle confinement within PIM as well as decreased the permeation of cyclosporine A and ovalbumin through PIM. Surprisingly, the viscosity of PGM and the permeation of cyclosporine A and ovalbumin through PGM was unaffected by the presence of SNAC, thus the effect of SNAC depended on the regional site that mucus was collected from. In the absence of SNAC, the permeation of different molecular weight and differently charged FDs through PIM was comparable to that through BM. However, while bulk permeation of neither of the FDs through PIM was affected by SNAC, the presence of SNAC decreased the permeation of FD4 and increased the permeation of FD150 kDa through BM. Additionally, and in contrast to observations in PIM, nanoparticle confinement within BM remained unaffected by the presence of SNAC. In conclusion, the present study showed that SNAC altered the physical and barrier properties of PIM, but not of PGM. The effects of SNAC in PIM were not observed in the BM in vitro model. Altogether, the study highlights the need for further understanding how permeation enhancers influence the mucus barrier and illustrates that the selected mucus model for such studies should be chosen with care.
Collapse
Affiliation(s)
- J S Mortensen
- Center for Biopharmaceuticals and Biobarriers in Drug Delivery, Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark
| | - S S-R Bohr
- Center for Biopharmaceuticals and Biobarriers in Drug Delivery, Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark; Department of Chemistry, Nano-Science Center, Faculty of Science, University of Copenhagen, Bülowsvej 17, DK-1870 Frederiksberg, Denmark
| | - S Harloff-Helleberg
- Center for Biopharmaceuticals and Biobarriers in Drug Delivery, Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark; LEO Foundation Center for Cutaneous Drug Delivery, Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark
| | - N S Hatzakis
- Department of Chemistry, Nano-Science Center, Faculty of Science, University of Copenhagen, Bülowsvej 17, DK-1870 Frederiksberg, Denmark; Novo Nordisk Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, DK-2200 Copenhagen, Denmark
| | - L Saaby
- CNS Drug Delivery and Barrier Modelling, Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark; Bioneer A/S, Kogle Alle 2, DK-2970 Hørsholm, Denmark
| | - H M Nielsen
- Center for Biopharmaceuticals and Biobarriers in Drug Delivery, Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark.
| |
Collapse
|
14
|
Ruiz-Pulido G, Quintanar-Guerrero D, Serrano-Mora LE, Medina DI. Triborheological Analysis of Reconstituted Gastrointestinal Mucus/Chitosan:TPP Nanoparticles System to Study Mucoadhesion Phenomenon under Different pH Conditions. Polymers (Basel) 2022; 14:4978. [PMID: 36433107 PMCID: PMC9696252 DOI: 10.3390/polym14224978] [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: 10/27/2022] [Revised: 11/07/2022] [Accepted: 11/08/2022] [Indexed: 11/19/2022] Open
Abstract
Polymeric nanoparticles have attracted much attention as pharmaceutical delivery vehicles to prolong residence time and enhance the bioavailability of therapeutic molecules through the mucoadhesive phenomenon. In this study, chitosan:TPP nanoparticles were synthetized using the ionic gelation technique to analyze their mucoadhesive interaction with reconstituted porcine gastrointestinal mucus from a triborheological point of view under different pH conditions (pH = 2.0, 4.0, 6.0 and 7.0). The triborheological profile of the reconstituted mucus was evaluated at different pH environments through the oscillation frequency and the flow sweep tests, demonstrating that the reconstituted mucus exhibits shear thinning behavior regardless of pH, while its viscoelastic properties showed a change in behavior from a polymeric solution performance under neutral pH conditions to a viscoelastic gel under acidic conditions. Additionally, a rheological synergism analysis was performed to visualize the changes that occur in the viscoelastic properties, the viscosity and the coefficient of friction of the reconstituted mucus samples as a consequence of the interaction with the chitosan:TPP nanoparticles to determine or to discard the presence of the mucoadhesion phenomenon under the different pH values. Mucoadhesiveness evaluation revealed that chitosan:TPP exhibited strong mucoadhesion under highly acidic pH conditions, below its pKa value of 6.5. In contrast, at neutral conditions or close to its pKa value, the chitosan:TPP nanoparticles' mucoadhesiveness was negligible.
Collapse
Affiliation(s)
- Gustavo Ruiz-Pulido
- Tecnologico de Monterrey, School of Engineering and Sciences, Atizapan de Zaragoza 52926, Estado de México, Mexico
| | - David Quintanar-Guerrero
- Laboratorio de Investigación y Posgrado en Tecnología Farmacéutica, FES-Cuautitlán, Universidad Nacional Autónoma de México, Cuautitlán Izcalli 54745, Estado de México, Mexico
| | - Luis Eduardo Serrano-Mora
- Laboratorio de Investigación y Posgrado en Tecnología Farmacéutica, FES-Cuautitlán, Universidad Nacional Autónoma de México, Cuautitlán Izcalli 54745, Estado de México, Mexico
| | - Dora I. Medina
- Tecnologico de Monterrey, Institute of Advanced Materials for Sustainable Manufacturing, Monterrey 64849, Nuevo Leon, Mexico
| |
Collapse
|
15
|
Kalia VC, Shim WY, Patel SKS, Gong C, Lee JK. Recent developments in antimicrobial growth promoters in chicken health: Opportunities and challenges. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 834:155300. [PMID: 35447189 DOI: 10.1016/j.scitotenv.2022.155300] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 04/04/2022] [Accepted: 04/11/2022] [Indexed: 06/14/2023]
Abstract
With a continuously increasing human population is an increasing global demand for food. People in countries with a higher socioeconomic status tend to switch their preferences from grains to meat and high-value foods. Their preference for chicken as a source of protein has grown by 70% over the last three decades. Many studies have shown the role of feed in regulating the animal gut microbiome and its impact on host health. The microbiome absorbs nutrients, digests foods, induces a mucosal immune response, maintains homeostasis, and regulates bioactive metabolites. These metabolic activities are influenced by the microbiota and diet. An imbalance in microbiota affects host physiology and progressively causes disorders and diseases. With the use of antibiotics, a shift from dysbiosis with a higher density of pathogens to homeostasis can occur. However, the progressive use of higher doses of antibiotics proved harmful and resulted in the emergence of multidrug-resistant microbes. As a result, the use of antibiotics as feed additives has been banned. Researchers, regulatory authorities, and managers in the poultry industry have assessed the challenges associated with these restrictions. Research has sought to identify alternatives to antibiotic growth promoters for poultry that do not have any adverse effects. Modulating the host intestinal microbiome by regulating dietary factors is much easier than manipulating host genetics. Research efforts have led to the identification of feed additives, including bacteriocins, immunostimulants, organic acids, phytogenics, prebiotics, probiotics, phytoncides, and bacteriophages. In contrast to focusing on one or more of these alternative bioadditives, an improved feed conversion ratio with enhanced poultry products is possible by employing a combination of feed additives. This article may be helpful in future research towards developing a sustainable poultry industry through the use of the proposed alternatives.
Collapse
Affiliation(s)
- Vipin Chandra Kalia
- Department of Chemical Engineering, Konkuk University, 1 Hwayang-Dong, Gwangjin-Gu, Seoul 05029, Republic of Korea.
| | - Woo Yong Shim
- Samsung Particulate Matter Research Institute, Samsung Advanced Institute of Technology (SAIT), Samsung Electronics Co., Ltd., 130 Samsung-ro, Yeongtong-gu, Suwon-si, Gyeonggi-do 16678, Republic of Korea
| | - Sanjay Kumar Singh Patel
- Department of Chemical Engineering, Konkuk University, 1 Hwayang-Dong, Gwangjin-Gu, Seoul 05029, Republic of Korea
| | - Chunjie Gong
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan 430068, People's Republic of China
| | - Jung-Kul Lee
- Department of Chemical Engineering, Konkuk University, 1 Hwayang-Dong, Gwangjin-Gu, Seoul 05029, Republic of Korea.
| |
Collapse
|
16
|
Senevirathne A, Hewawaduge C, Lee JH. Assessing an O-antigen deficient, live attenuated Salmonella Gallinarium strain that is DIVA compatible, environmentally safe, and protects chickens against fowl typhoid. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2022; 133:104433. [PMID: 35568244 DOI: 10.1016/j.dci.2022.104433] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 04/25/2022] [Accepted: 05/06/2022] [Indexed: 06/15/2023]
Abstract
The objective of the present study was to create a highly attenuated, safe Salmonella Gallinarium (SG) vaccine strain for chicken vaccination against fowl typhoid (FT) diseases. The SG vaccine strain (SGVS) consists of three virulence-related gene deletions, namely, lon, cpxR, and rfaL. The parent strain (SGPS) with Δlon ΔcpxR genotype was utilized as the host strain for in-frame rfaL gene deletion by lambda red recombination. The SGVS was highly attenuated with improved environmental safety by zero fecal contamination beyond seven days for both oral and intramuscular immunization routes. Upon inoculation into 1-month-old young chicken, no vaccine-induced adverse behaviors were observed and did not cause a chronic state of infection as the SG wild-type strain did. Immunization of chicken elicited both humoral and cell-mediated immune responses demarcated by, IgY antibody assessment, T-cell responses in peripheral blood mononuclear cells, and the induction of immunomodulatory cytokines, IFN-γ, IL-2, IL-12, and IL-4 to resemble both Th1 and Th2 type of immune responses. The immunological assessment revealed a high level of efficacy of the SGVS when inoculated via the IM route than the oral route. The strain was less cytotoxic with reduced cytotoxicity on chicken macrophages and was DIVA capable with minimum reactivity of immunized serum with purified SG lipopolysaccharides. The challenge study could generate 70% protection in chicken for SGVS, whereas no birds were protected in the PBS challenged group. The protection levels were evident in histopathological assessment of spleen and liver specimens and also the external appearance of the spleen with reduced lesions on immunized groups. Further experiments may be warranted to dose and route optimization for further increase in the protection level derived by present SGVS.
Collapse
Affiliation(s)
- Amal Senevirathne
- College of Veterinary Medicine, Jeonbuk National University, Iksan Campus, 54596, South Korea
| | - Chamith Hewawaduge
- College of Veterinary Medicine, Jeonbuk National University, Iksan Campus, 54596, South Korea
| | - John Hwa Lee
- College of Veterinary Medicine, Jeonbuk National University, Iksan Campus, 54596, South Korea.
| |
Collapse
|
17
|
Huang X, Liu S, Zuo F, Luo L, Chen D, Ou Y, Geng Y, Zhang Y, Lin G, Yang S, Luo W, Yin L, He Z. cMOS enhanced the mucosal immune function of skin and gill of goldfish (Carassius auratus Linnaeus) to improve the resistance to Ichthyophthirius multifiliis infection. FISH & SHELLFISH IMMUNOLOGY 2022; 126:1-11. [PMID: 35595060 DOI: 10.1016/j.fsi.2022.05.024] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 05/10/2022] [Accepted: 05/12/2022] [Indexed: 06/15/2023]
Abstract
of supporting mucosal immune barrier integrity and prevention of some pathogenic infections in aquatic species, are key areas of active study, often focusing on feed additives. The objectives of this study were to explore the effects of feeding cMOS (concentrated mannan oligosaccharide) on the gill and skin mucosal barriers of goldfish (Carassius auratus Linnaeus) and evaluate health status during Ichthyophthirius multifiliis infection. After feeding the cMOS-containing diet for 60 days, Hematoxylin and eosin (H&E) staining showed greater length of gill lamella and thicker dermal dense layer, while Alcian Blue and Periodic acid-Schiff (AB-PAS) staining showed higher numbers of mucin cells in cMOS fed fish. Chemical analysis showed that fish fed cMOS had greater enzyme activity of lysozyme (LZM) and alkaline phosphatase (AKP) in gill and skin tissues, while qRT-PCR revealed higher expression of Muc-2 and IL-1β, as well as lower expression of IL-10. After Ichthyophthirius multifiliis challenge, goldfish fed the cMOS diet had lower mortality and infection rates, as well as fewer visible white spots on the body surfaces. Histologically, the gill and skin of these fish presented less tissue damage and fewer parasites, and had a greater number of mucus cells. In addition, the expression of Muc-2 and IL-10 were notably higher while the expression of IL-1β was significantly lower in cMOS fed goldfish than control fed fish. In this study, cMOS fed goldfish had stronger immune barrier function of skin and gill mucous, and better survival following Ichthyophthirius multifiliis infection.
Collapse
Affiliation(s)
- Xiaoli Huang
- Department of Aquaculture, College of Animal Science & Technology, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Senyue Liu
- Department of Aquaculture, College of Animal Science & Technology, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Fengyuan Zuo
- Department of Aquaculture, College of Animal Science & Technology, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Lin Luo
- Department of Aquaculture, College of Animal Science & Technology, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Defang Chen
- Department of Aquaculture, College of Animal Science & Technology, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Yangping Ou
- Department of Basic Veterinary, College of Veterinary Medicine, Sichuan Agricultural University, Chendu, 611130, Sichuan, China
| | - Yi Geng
- Department of Basic Veterinary, College of Veterinary Medicine, Sichuan Agricultural University, Chendu, 611130, Sichuan, China.
| | - Yufan Zhang
- Alltech Biological Products (China) Co. Ltd, 100060, Beijing, China
| | - Gang Lin
- Alltech Biological Products (China) Co. Ltd, 100060, Beijing, China
| | - Shiyong Yang
- Department of Aquaculture, College of Animal Science & Technology, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Wei Luo
- Department of Aquaculture, College of Animal Science & Technology, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Lizi Yin
- Department of Basic Veterinary, College of Veterinary Medicine, Sichuan Agricultural University, Chendu, 611130, Sichuan, China
| | - Zhi He
- Department of Aquaculture, College of Animal Science & Technology, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| |
Collapse
|
18
|
Kirthika P, Park S, Jawalagatti V, Lee JH. Evaluation of host and bacterial gene modulation during Lawsonia intracellularis infection in immunocompetent C57BL/6 mouse model. J Vet Sci 2022; 23:e41. [PMID: 35332712 PMCID: PMC9149498 DOI: 10.4142/jvs.21274] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 01/02/2022] [Accepted: 01/27/2022] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Proliferative enteritis caused by Lawsonia intracellularis undermines the economic stability of the swine industry worldwide. The development of cost-effective animal models to study the pathophysiology of the disease will help develop strategies to counter this bacterium. OBJECTIVES This study focused on establishing a model of gastrointestinal (GI) infection of L. intracellularis in C57BL/6 mice to evaluate the disease progression and lesions of proliferative enteropathy (PE) in murine GI tissue. METHODS We assessed the murine mucosal and cell-mediated immune responses generated in response to inoculation with L. intracellularis. RESULTS The mice developed characteristic lesions of the disease and shed L. intracellularis in the feces following oral inoculation with 5 × l07 bacteria. An increase in L. intracellularis 16s rRNA and groEL copies in the intestine of infected mice indicated intestinal dissemination of the bacteria. The C57BL/6 mice appeared capable of modulating humoral and cell-mediated immune responses to L. intracellularis infection. Notably, the expression of genes for the vitamin B12 receptor and for secreted and membrane-bound mucins were downregulated in L. intracellularis -infected mice. Furthermore, L. intracellularis colonization of the mouse intestine was confirmed by the immunohistochemistry and western blot analyses. CONCLUSIONS This is the first study demonstrating the contributions of bacterial chaperonin and host nutrient genes to PE using an immunocompetent mouse model. This mouse infection model may serve as a platform from which to study L. intracellularis infection and develop potential vaccination and therapeutic strategies to treat PE.
Collapse
Affiliation(s)
| | - Sungwoo Park
- College of Veterinary Medicine, Jeonbuk National University, Iksan 54596, Korea
| | | | - John Hwa Lee
- College of Veterinary Medicine, Jeonbuk National University, Iksan 54596, Korea.
| |
Collapse
|
19
|
Padra JT, Loibman SO, Thorell K, Sundh H, Sundell K, Lindén SK. Atlantic Salmon Mucins Inhibit LuxS-Dependent A. Salmonicida AI-2 Quorum Sensing in an N-Acetylneuraminic Acid-Dependent Manner. Int J Mol Sci 2022; 23:ijms23084326. [PMID: 35457143 PMCID: PMC9026418 DOI: 10.3390/ijms23084326] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 04/11/2022] [Accepted: 04/12/2022] [Indexed: 02/06/2023] Open
Abstract
One of the most important bacterial diseases in salmonid aquaculture is furunculosis, caused by Aeromonas salmonicida. Bacterial communication through secreted autoinducer signals, quorum sensing, takes part in the regulation of gene expression in bacteria, influencing growth and virulence. The skin and mucosal surfaces, covered by a mucus layer, are the first point of contact between fish and bacteria. Mucins are highly glycosylated and are the main components of mucus. Here, we validate the Vibrio harveyi BB170 bioreporter assay for quantifying A. salmonicida quorum sensing and study the effects of Atlantic salmon mucins as well as mono- and disaccharides on the AI-2 levels of A. salmonicida. Atlantic salmon mucins from skin, pyloric ceca, proximal and distal intestine reduced A. salmonicida AI-2 levels. Among the saccharides abundant on mucins, fucose, N-acetylneuraminic acid and GlcNAcβ1-3Gal inhibited AI-2 A. salmonicida secretion. Removal of N-acetylneuraminic acid, which is the most abundant terminal residue on mucin glycans on Atlantic salmon mucins, attenuated the inhibitory effects on AI-2 levels of A. salmonicida. Deletion of A. salmonicida luxS abolished AI-2 production. In conclusion, Atlantic salmon mucins regulate A. salmonicida quorum sensing in a luxS and N-acetylneuraminic acid-dependent manner.
Collapse
Affiliation(s)
- János Tamás Padra
- Department of Medical Chemistry and Cell Biology, University of Gothenburg, 405 30 Gothenburg, Sweden; (J.T.P.); (S.O.L.); (K.T.)
| | - Stefany Ojaimi Loibman
- Department of Medical Chemistry and Cell Biology, University of Gothenburg, 405 30 Gothenburg, Sweden; (J.T.P.); (S.O.L.); (K.T.)
| | - Kaisa Thorell
- Department of Medical Chemistry and Cell Biology, University of Gothenburg, 405 30 Gothenburg, Sweden; (J.T.P.); (S.O.L.); (K.T.)
| | - Henrik Sundh
- Department of Biological and Environmental Sciences, University of Gothenburg, 405 30 Gothenburg, Sweden; (H.S.); (K.S.)
| | - Kristina Sundell
- Department of Biological and Environmental Sciences, University of Gothenburg, 405 30 Gothenburg, Sweden; (H.S.); (K.S.)
| | - Sara K. Lindén
- Department of Medical Chemistry and Cell Biology, University of Gothenburg, 405 30 Gothenburg, Sweden; (J.T.P.); (S.O.L.); (K.T.)
- Correspondence:
| |
Collapse
|
20
|
Yang WY, Chou CH, Wang C. The Effects of Feed Supplementing Akkemansia muciniphila on Incidence, Severity, and Gut Microbiota of Necrotic Enteritis in Chickens. Poult Sci 2022; 101:101751. [PMID: 35240353 PMCID: PMC8889413 DOI: 10.1016/j.psj.2022.101751] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 01/06/2022] [Accepted: 01/25/2022] [Indexed: 01/07/2023] Open
Abstract
Akkermansia muciniphila (AM) is a mucin-degrading anaerobe, exerting beneficial effects on gut integrity improvement, inflammatory alleviation, and metabolic regulations in humans. Excess amounts of mucin and mucogenesis in the gut facilitate the development of necrotic enteritis (NE) in chickens. The study aimed to evaluate the effects of oral inoculation of AM on NE prevention and gut modulation in a NE-reproduced model coinfecting with Clostridium perfringens (CP) and Eimeria parasites. A total of 105 commercial 1-day-old broilers were randomly allocated into 5 groups, respectively challenged with Eimeria (Eimeria group), Eimeria and CP (Eimeria+CP group), Eimeria and CP with AM (Eimeria+CP+AM group), Eimeria and AM (Eimeria+AM group), and a placebo (Noninfected group). The treatment of AM exhibited a low degree of amelioration on NE severity. The application neither protected broilers from NE by decreasing NE-positive numbers nor reached a significant reduction in lesion scores in the small intestines. The development of NE reduced species diversity in jejunal microbiota; the pretreatments of AM exacerbated the consequence by losing species richness and promoted the similarity of the jejunal microbial community presented in the Eimeria+CP group. The participation of AM enhanced the increments of genera Clostridium sensu stricto 1 and Escherichia_Shigella and decreased the number of Lactobacillus. The significant variations of genera Clostridium sensu stricto 1 and Lactobacillus in jejunal microbiota were associated with NE development and promotion. In conclusion, oral inoculation of AM promoted the development of NE and modulated the jejunal microbiota favorable for CP overgrowth in broilers. The application of AM as a probiotic in broilers should be cautious on account of the effects to predispose NE.
Collapse
Affiliation(s)
- Wen-Yuan Yang
- Department of Veterinary Medicine, School of Veterinary Medicine, National Taiwan University, Taipei City 106, Taiwan; Zoonoses Research Center and School of Veterinary Medicine, National Taiwan University, Taipei City, 106, Taiwan
| | - Chung-Hsi Chou
- Department of Veterinary Medicine, School of Veterinary Medicine, National Taiwan University, Taipei City 106, Taiwan; Zoonoses Research Center and School of Veterinary Medicine, National Taiwan University, Taipei City, 106, Taiwan
| | - Chinling Wang
- Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, MS 39762, USA.
| |
Collapse
|
21
|
Brachyspira Species Avidity to Colonic Mucins from Pigs with and without Brachyspira hyodysenteriae Infection Is Species Specific and Varies between Strains. Infect Immun 2021; 89:e0048621. [PMID: 34543117 DOI: 10.1128/iai.00486-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Brachyspira hyodysenteriae is commonly associated with swine dysentery (SD), a disease that has an economic impact on the swine industry. B. hyodysenteriae infection results in changes to the colonic mucus niche with massive mucus induction, which substantially increases the number of B. hyodysenteriae binding sites in the mucus. We previously determined that a B. hyodysenteriae strain binds to colon mucins in a manner that differs between pigs and mucin types. Here, we investigated if adhesion to mucins is a trait observed across a broad set of B. hyodysenteriae strains and isolates and furthermore at a genus level (B. innocens, B. pilosicoli, B. murdochii, B. hampsonii, and B. intermedia strains). Our results show that binding to mucins appears to be specific to B. hyodysenteriae, and within this species, the binding ability to mucins varies between strains/isolates, increases for mucins from pigs with SD, and is associated with sialic acid epitopes on mucins. Infection with B. hyodysenteriae strain 8dII results in mucin glycosylation changes in the colon, including a shift in sialic acid-containing structures. Thus, we demonstrate through hierarchical cluster analysis and orthogonal projections to latent structures discriminant analysis (OPLS-DA) models of the relative abundances of sialic acid-containing glycans that sialic acid-containing structures in the mucin O-glycome are good predictors of B. hyodysenteriae strain 8dII infection in pigs. The results emphasize the role of sialic acids in governing B. hyodysenteriae interactions with its host, which may open perspectives for therapeutic strategies.
Collapse
|
22
|
Lima PC, Hartley-Tassell L, Wynne JW. The ability of Neoparamoeba perurans to bind to and digest non-fish-derived mucin: Insights into the amoeba's mechanism of action to overcome gill mucus production. JOURNAL OF FISH DISEASES 2021; 44:1355-1367. [PMID: 33990985 DOI: 10.1111/jfd.13394] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 04/22/2021] [Accepted: 04/26/2021] [Indexed: 06/12/2023]
Abstract
Amoebic gill disease (AGD) is caused by the marine amoeba Neoparamoeba perurans, a facultative parasite. Despite the significant impact this disease has on production of Atlantic salmon worldwide, the mechanisms involved in host-parasite interaction remains unknown. Excessive gill mucus secretion is reported as a host defence mechanism to prevent microbial colonization in the gill epithelium. Despite this response, N. perurans still attaches and proliferates. The present study aimed to investigate the interaction between N. perurans and mucin, the most abundant component in mucus. An in vitro adhesion assay using bovine submaxillary mucin (BSM) demonstrated that amoeba binding to mucin-coated substrate was significantly higher than to the BSA control. This binding interaction is likely glycan-mediated as pre-incubation with galactose, galactosamine, N-acetylgalactosamine and fucose reduced mucin adhesion to control levels. The ability of N. perurans to secrete proteases that target mucin was also investigated. Protease activity was detected in the amoeba culture media in the presence of BSM, but not when protease inhibitor was added. Mucin degradation was visually assessed on protein gels. This study provides preliminary evidence that N. perurans has developed mechanisms to interact with and evade mucus by binding to mucin glycan receptors and secreting proteases with mucolytic activity.
Collapse
Affiliation(s)
- Paula C Lima
- CSIRO Livestock & Aquaculture Program, Queensland, Australia
| | | | - James W Wynne
- CSIRO Livestock & Aquaculture Program, Tasmania, Australia
| |
Collapse
|
23
|
Padra JT, Lindén SK. Optimization of Alcian blue pH 1.0 histo-staining protocols to match mass spectrometric quantification of sulfomucins and circumvent false positive results due to sialomucins. Glycobiology 2021; 32:6-10. [PMID: 34420054 PMCID: PMC8881734 DOI: 10.1093/glycob/cwab091] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Revised: 08/10/2021] [Accepted: 08/10/2021] [Indexed: 11/16/2022] Open
Abstract
Sulfomucins are in some body locations and species a normal occurrence, whereas in other situations, are a sign of pathology. Sulfomucin content on histological sections and isolated material is frequently analyzed with Alcian blue staining at pH 1.0. However, since the stain detects the charge, a high density of other charged molecules, such as sialic acids, has potential to impede specificity. Here, we compared the outcome from four staining protocols with the level of sulfation determined by liquid chromatography–tandem mass spectrometric analysis on samples from various tissues with variable sulfation and sialylation levels. We found that a protocol we designed, including rinsing with MetOH and 0.5 M NaCl buffer at pH 1.0, eliminates the false positive staining of tissues outperforming commonly recommended solutions. In tissues with low-to-moderately sulfated mucins (e.g. human stomach and salmonid epithelia), this method enables accurate relative quantification (e.g. sulfate scoring comparisons between healthy and diseased tissues), whereas the range of the method is not suitable for comparisons between tissues with high sulfomucin content (e.g. pig stomach and colon).
Collapse
Affiliation(s)
- János Tamás Padra
- Department of Medical Chemistry and Cell Biology, University of Gothenburg, Gothenburg, Sweden
| | - Sara K Lindén
- Department of Medical Chemistry and Cell Biology, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
24
|
Kumar V, Roy S, Behera BK, Bossier P, Das BK. Acute Hepatopancreatic Necrosis Disease (AHPND): Virulence, Pathogenesis and Mitigation Strategies in Shrimp Aquaculture. Toxins (Basel) 2021; 13:524. [PMID: 34437395 PMCID: PMC8402356 DOI: 10.3390/toxins13080524] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 07/20/2021] [Accepted: 07/22/2021] [Indexed: 02/06/2023] Open
Abstract
Shrimp, as a high-protein animal food commodity, are one of the fastest growing food producing sectors in the world. It has emerged as a highly traded seafood product, currently exceeding 8 MT of high value. However, disease outbreaks, which are considered as the primary cause of production loss in shrimp farming, have moved to the forefront in recent years and brought socio-economic and environmental unsustainability to the shrimp aquaculture industry. Acute hepatopancreatic necrosis disease (AHPND), caused by Vibrio spp., is a relatively new farmed penaeid shrimp bacterial disease. The shrimp production in AHPND affected regions has dropped to ~60%, and the disease has caused a global loss of USD 43 billion to the shrimp farming industry. The conventional approaches, such as antibiotics and disinfectants, often applied for the mitigation or cure of AHPND, have had limited success. Additionally, their usage has been associated with alteration of host gut microbiota and immunity and development of antibiotic resistance in bacterial pathogens. For example, the Mexico AHPND-causing V. parahaemolyticus strain (13-306D/4 and 13-511/A1) were reported to carry tetB gene coding for tetracycline resistance gene, and V. campbellii from China was found to carry multiple antibiotic resistance genes. As a consequence, there is an urgent need to thoroughly understand the virulence mechanism of AHPND-causing Vibrio spp. and develop novel management strategies to control AHPND in shrimp aquaculture, that will be crucially important to ensure food security in the future and offer economic stability to farmers. In this review, the most important findings of AHPND are highlighted, discussed and put in perspective, and some directions for future research are presented.
Collapse
Affiliation(s)
- Vikash Kumar
- Aquatic Environmental Biotechnology and Nanotechnology (AEBN) Division, ICAR-Central Inland Fisheries Research Institute (CIFRI), Barrackpore 700120, India; (S.R.); (B.K.B.); (B.K.D.)
- Laboratory of Aquaculture & Artemia Reference Center, Department of Animal Sciences and Aquatic Ecology, Faculty of Bioscience Engineering, Ghent University, B-9000 Ghent, Belgium;
| | - Suvra Roy
- Aquatic Environmental Biotechnology and Nanotechnology (AEBN) Division, ICAR-Central Inland Fisheries Research Institute (CIFRI), Barrackpore 700120, India; (S.R.); (B.K.B.); (B.K.D.)
- Laboratory of Aquaculture & Artemia Reference Center, Department of Animal Sciences and Aquatic Ecology, Faculty of Bioscience Engineering, Ghent University, B-9000 Ghent, Belgium;
| | - Bijay Kumar Behera
- Aquatic Environmental Biotechnology and Nanotechnology (AEBN) Division, ICAR-Central Inland Fisheries Research Institute (CIFRI), Barrackpore 700120, India; (S.R.); (B.K.B.); (B.K.D.)
| | - Peter Bossier
- Laboratory of Aquaculture & Artemia Reference Center, Department of Animal Sciences and Aquatic Ecology, Faculty of Bioscience Engineering, Ghent University, B-9000 Ghent, Belgium;
| | - Basanta Kumar Das
- Aquatic Environmental Biotechnology and Nanotechnology (AEBN) Division, ICAR-Central Inland Fisheries Research Institute (CIFRI), Barrackpore 700120, India; (S.R.); (B.K.B.); (B.K.D.)
| |
Collapse
|
25
|
Rahardja F, Prasetyo D, Shahib MN, Tjahjani S. The Influence of Lactobacillus Acidophilus on MUC1, GAL-3, IL-1β and IL-17 Gene Expression in BALB/c Mice Stomach. Open Microbiol J 2021. [DOI: 10.2174/1874285802115010067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Background and Objective:
Lactobacillus acidophilus has been widely used for the management of gastrointestinal carcinoma owing to its immunomodulation effect; however, the role of L. acidophilus and its specific mechanism of action in the stomach is not fully comprehended. The present study evaluated the expression profile of MUC-1, GAL-3, IL -1β, and IL-17 in the L. acidophilus treated mice stomach.
Methods:
The study was conducted utilizing three groups of mice, 6 mice for each group, administered with different doses of L. acidophilus and a control group treated with normal saline. The results were analyzed with the Mann-Whitney Test.
Results:
The results demonstrated that L. acidophilus elevated IL-1β insignificantly and inhibited the expression of IL-17. The MUC-1 expression is influenced by L. acidophilus and inversely proportional to GAL-3 expression.
Conclusion:
Lactobacillus acidophilus plays a prominent role against inflammatory responses and has a potential in the treatment of gastrointestinal cancer.
Collapse
|
26
|
Du E, Guo Y. Dietary supplementation of essential oils and lysozyme reduces mortality and improves intestinal integrity of broiler chickens with necrotic enteritis. Anim Sci J 2021; 92:e13499. [PMID: 33455052 DOI: 10.1111/asj.13499] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 09/29/2020] [Accepted: 10/15/2020] [Indexed: 01/08/2023]
Abstract
The objective of this study was to investigate the individual and combined effects of essential oils (EO; comprised of thymol and carvacrol) and lysozyme on experimental NE in broiler chickens. A total of 320 1-day-old chicks were randomly assigned to five treatment groups: no-challenge control (NC), NC + C. perfringens challenge (CC), CC + 120 mg/kg of EO, CC + 100 mg/kg of lysozyme, and CC + 120 mg/kg of EO + 100 mg/kg of lysozyme. The results showed that EO or lysozyme decreased the mortality, alleviated the gut lesions, inhibited the liver Enterobacteriaceae carriage, and increased the villus height of the ileum compared with CC (p < .05), although the proliferation of C. perfringens in the ileum was not inhibited (p > .05). Moreover, EO or lysozyme was found to decrease the ileal concentration of sialic acid and the Mucin2 mRNA expression (p < .05). However, the blend of EO and lysozyme did not display significant effect on the NE-associated mortality or gut damage in contrast with CC (p > .05). In conclusion, these findings suggest the similar protective effects of EO and lysozyme in NE-associated mortality and intestinal impairment, but their blend did not exhibit ameliorative effect.
Collapse
Affiliation(s)
- Encun Du
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, P. R. China.,Hubei Key Laboratory of Animal Embryo and Molecular Breeding, Institute of Animal Science and Veterinary Medicine, Hubei Academy of Agricultural Sciences, Wuhan, P. R. China
| | - Yuming Guo
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, P. R. China
| |
Collapse
|
27
|
Gautier T, David-Le Gall S, Sweidan A, Tamanai-Shacoori Z, Jolivet-Gougeon A, Loréal O, Bousarghin L. Next-Generation Probiotics and Their Metabolites in COVID-19. Microorganisms 2021; 9:microorganisms9050941. [PMID: 33925715 PMCID: PMC8146258 DOI: 10.3390/microorganisms9050941] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 04/24/2021] [Accepted: 04/26/2021] [Indexed: 02/06/2023] Open
Abstract
Since December 2019, a global pandemic has been observed, caused by the emergence of a new coronavirus, SARS CoV-2. The latter is responsible for the respiratory disease, COVID-19. The infection is also characterized by renal, hepatic, and gastrointestinal dysfunctions suggesting the spread of the virus to other organs. A dysregulated immune response was also reported. To date, there is no measure to treat or prevent SARS CoV-2 infection. Additionally, as gut microbiota composition is altered in patients with COVID-19, alternative therapies using probiotics can be considered to fight SARS CoV-2 infection. This review aims at summarizing the current knowledge about next-generation probiotics (NGPs) and their benefits in viral respiratory tract infections and in COVID-19. We describe these bacteria, highlighted by studies using metagenomic approaches. In addition, these bacteria generate metabolites such as butyrate, desaminotyrosine, and secondary bile acid, suggested to prevent viral respiratory infections. Gut microbial metabolites transported via the circulation to the lungs could inhibit viral replication or improve the immune response against viruses. The use of probiotics and/or their metabolites may target either the virus itself and/or the immunologic process. However, this review showed that more studies are needed to determine the benefits of probiotics and metabolite products in COVID-19.
Collapse
Affiliation(s)
- Thomas Gautier
- UMR 1241, Nutrition Metabolisms and Cancer Institute, Inserm, INRAE, Université de Rennes 1, 35000 Rennes, France; (T.G.); (S.D.-L.G.); (Z.T.-S.); (A.J.-G.); (O.L.)
| | - Sandrine David-Le Gall
- UMR 1241, Nutrition Metabolisms and Cancer Institute, Inserm, INRAE, Université de Rennes 1, 35000 Rennes, France; (T.G.); (S.D.-L.G.); (Z.T.-S.); (A.J.-G.); (O.L.)
| | - Alaa Sweidan
- Laboratory of Microbiology, Department of Life and Earth Sciences, Faculty of Sciences I, Lebanese University, Hadath Campus, P.O. Box 6573/14 Beirut, Lebanon;
| | - Zohreh Tamanai-Shacoori
- UMR 1241, Nutrition Metabolisms and Cancer Institute, Inserm, INRAE, Université de Rennes 1, 35000 Rennes, France; (T.G.); (S.D.-L.G.); (Z.T.-S.); (A.J.-G.); (O.L.)
| | - Anne Jolivet-Gougeon
- UMR 1241, Nutrition Metabolisms and Cancer Institute, Inserm, INRAE, Université de Rennes 1, 35000 Rennes, France; (T.G.); (S.D.-L.G.); (Z.T.-S.); (A.J.-G.); (O.L.)
| | - Olivier Loréal
- UMR 1241, Nutrition Metabolisms and Cancer Institute, Inserm, INRAE, Université de Rennes 1, 35000 Rennes, France; (T.G.); (S.D.-L.G.); (Z.T.-S.); (A.J.-G.); (O.L.)
| | - Latifa Bousarghin
- UMR 1241, Nutrition Metabolisms and Cancer Institute, Inserm, INRAE, Université de Rennes 1, 35000 Rennes, France; (T.G.); (S.D.-L.G.); (Z.T.-S.); (A.J.-G.); (O.L.)
- Correspondence: ; Tel.: +33-2232-3489-8
| |
Collapse
|
28
|
Tran PTN, Kumar V, Bossier P. Do acute hepatopancreatic necrosis disease-causing PirAB VP toxins aggravate vibriosis? Emerg Microbes Infect 2021; 9:1919-1932. [PMID: 32799621 PMCID: PMC8284973 DOI: 10.1080/22221751.2020.1811778] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Gram-negative marine bacterium Vibrio parahaemolyticus is an important aquatic pathogen and has been demonstrated to be the causative agent of acute hepatopancreatic necrotic disease (AHPND) in shrimp aquaculture. The AHPND-causing V. parahaemolyticus strains contain a pVA1 plasmid encoding the binary PirAVP and PirBVP toxins, are the primary virulence factor that mediates AHPND and mortality in shrimp. Since PirABVP toxins are secreted extracellularly, one can hypothesize that PirABVP toxins would aggravate vibriosis in the aquatic environment. To address this, in vivo and in vitro experiments were conducted. Germ-free Artemia franciscana were co-challenged with PirABVP toxins and 10 Vibrio spp. The in vivo results showed that PirABVP toxin interact synergistically with MM30 (a quorum sensing AI-2 deficient mutant) and V. alginolyticus AQ13-91, aggravating vibriosis. However, co-challenge by PirABVP toxins and V. campbellii LMG21363, V. parahaemolyticus CAIM170, V. proteolyticus LMG10942, and V. anguillarum NB10 worked antagonistically, increasing the survival of Artemia larvae. The in vitro results showed that the addition of PirABVP toxins significantly modulated the production of the virulence factors of studied Vibrio spp. Yet these in vitro results did not help to explain the in vivo results. Hence it appears that PirABVP toxins can aggravate vibriosis. However, the dynamics of interaction is strain dependent.
Collapse
Affiliation(s)
- Phuong Thi Ngoc Tran
- Lab of Aquaculture & Artemia Reference Center, Department of Animal Sciences and Aquatic Ecology, Faculty of Bioscience Engineering, Ghent University Ghent, Belgium
| | - Vikash Kumar
- Lab of Aquaculture & Artemia Reference Center, Department of Animal Sciences and Aquatic Ecology, Faculty of Bioscience Engineering, Ghent University Ghent, Belgium.,ICAR-Central Inland Fisheries Research Institute (CIFRI), Barrackpore, India
| | - Peter Bossier
- Lab of Aquaculture & Artemia Reference Center, Department of Animal Sciences and Aquatic Ecology, Faculty of Bioscience Engineering, Ghent University Ghent, Belgium
| |
Collapse
|
29
|
Comparison of Strategies for Isolating Anaerobic Bacteria from the Porcine Intestine. Appl Environ Microbiol 2021; 87:AEM.00088-21. [PMID: 33608289 DOI: 10.1128/aem.00088-21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 02/08/2021] [Indexed: 12/22/2022] Open
Abstract
The isolation of bacteria that represent the diversity of autochthonous taxa in the gastrointestinal tract is necessary to fully ascertain their function, but the majority of bacterial species inhabiting the intestines of mammals are fastidious and thus challenging to isolate. The goal of the current study was to isolate a diverse assemblage of anaerobic bacteria from the intestine of pigs as a model animal and to comparatively examine various novel and traditional isolation strategies. Methods used included long-term enrichments, direct plating, a modified ichip method, as well as ethanol and tyndallization treatments of samples to select for endospore-forming taxa. A total of 234 taxa (91 previously uncultured) comprising 80 genera and 7 phyla were isolated from mucosal and luminal samples from the ileum, cecum, ascending colon, and spiral colon removed from animals under anesthesia. The diversity of bacteria isolated from the large intestine was less than that detected by next-generation sequence analysis. Long-term enrichments yielded the greatest diversity of recovered bacteria (Shannon's index [SI] = 4.7). Methods designed to isolate endospore-forming bacteria produced the lowest diversity (SI ≤ 2.7), with tyndallization yielding lower diversity than the ethanol method. However, the isolation frequency of previously uncultured bacteria was highest for ethanol-treated samples (41.9%) and the ichip method (32.5%). The goal of recovering a diverse collection of enteric bacteria was achieved. Importantly, the study findings demonstrate that it is necessary to use a combination of methods in concert to isolate bacteria that are representative of the diversity within the intestines of mammals.IMPORTANCE This work determined that using a combination of anaerobic isolation methods is necessary to increase the diversity of bacteria recovered from the intestines of monogastric mammals. Direct plating methods have traditionally been used to isolate enteric bacteria, and recent methods (e.g., diffusion methods [i.e., ichip] or differential isolation of endospore-forming bacteria) have been suggested to be superior at increasing diversity, including the recovery of previously uncultured taxa. We showed that long-term enrichment of samples using a variety of media isolated the most diverse and novel bacteria. Application of the ichip method delivered a diversity of bacteria similar to those of enrichment and direct plating methods. Methods that selected for endospore-forming bacteria generated collections that differed in composition from those of other methods with reduced diversity. However, the ethanol treatment frequently isolated novel bacteria. By using a combination of methods in concert, a diverse collection of enteric bacteria was generated for ancillary experimentation.
Collapse
|
30
|
Poudel S, Zhang L, Tabler GT, Lin J, Zhai W. Effects of riboflavin and Bacillus subtilis on internal organ development and intestinal health of Ross 708 male broilers with or without coccidial challenge. Poult Sci 2021; 100:100973. [PMID: 33588345 PMCID: PMC7896149 DOI: 10.1016/j.psj.2020.12.070] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Revised: 12/22/2020] [Accepted: 12/26/2020] [Indexed: 11/28/2022] Open
Abstract
In a companion study, we found that inclusion of different doses of riboflavin affected growth performance of Ross 708 male broilers' responses to coccidial challenge (by 5 Eimeria spp on day 14 of age) and dietary Bacillus subtilis (B. subtilis) supplementation. The current study was conducted to further test whether supplementation of B. subtilis and riboflavin will reduce negative impact and inflammation caused by Eimeria spp proliferation and help proper function of internal organs. A total of 1,248 Ross × Ross 708 male broiler chicks were randomly placed in 96 floor pens (8 blocks, 12 treatments). Treatments were arranged in a 3 (riboflavin) × 2 (B. subtilis) × 2 (Coccidial challenge) factorial arrangement in a randomized complete block design. Coccidial challenge reduced the weight of sampled birds on day 27 and day 36 and increased the relative weights of the internal organs of proventriculus, duodenum, jejunum, ileum, and spleen to BW on day 27, which may be because of inflammation caused by proliferation of Eimeria spp. The increased relative weights of duodenum, jejunum, ileum, and spleen on coccidial challenged birds were lost on day 36. Correlation analysis also indicated that the jejunum weight was positively related to villus height, Eimeria acervulina, and Eimeria maxima on day 27 but was not on day 36. The loss of the positive relationships may be because of recovery of the birds from coccidiosis on day 36. Even though the coccidial challenge and riboflavin interactively affected feed conversion ratio and BW gain and supplementation of dietary B. subtilis reduced mortality from day 35 to 42 in the companion study, the same response of internal organs was not observed in the current study. Coccidial challenge compromised development of internal organs of Ross 708 male broilers at an early age, but the negative effects subsided with age of birds rather than supplementation of riboflavin and B. subtilis at current tested levels under our experimental set up.
Collapse
Affiliation(s)
- Sabin Poudel
- Department of Poultry Science, Mississippi State University, Mississippi State, MS, USA
| | - Li Zhang
- Department of Poultry Science, Mississippi State University, Mississippi State, MS, USA
| | - George T Tabler
- Department of Poultry Science, Mississippi State University, Mississippi State, MS, USA
| | - Jun Lin
- Department of Animal Science, University of Tennessee, Knoxville, TN, USA
| | - Wei Zhai
- Department of Poultry Science, Mississippi State University, Mississippi State, MS, USA.
| |
Collapse
|
31
|
Kikusato M. Phytobiotics to improve health and production of broiler chickens: functions beyond the antioxidant activity. Anim Biosci 2021; 34:345-353. [PMID: 33705621 PMCID: PMC7961201 DOI: 10.5713/ab.20.0842] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 02/02/2021] [Indexed: 12/17/2022] Open
Abstract
Phytobiotics, also known as phytochemicals or phytogenics, have a wide variety of biological activities and have recently emerged as alternatives to synthetic antibiotic growth promoters. Numerous studies have reported the growth-promoting effects of phytobiotics in chickens, but their precise mechanism of action is yet to be elucidated. Phytobiotics are traditionally known for their antioxidant activity. However, extensive investigations have shown that these compounds also have anti-inflammatory, antimicrobial, and transcription-modulating effects. Phytobiotics are non-nutritive constituents, and their bioavailability is low. Nonetheless, their beneficial effects have been observed in several tissues or organs. The health benefits of the ingestion of phytobiotics are attributed to their antioxidant activity. However, several studies have revealed that not all these benefits could be explained by the antioxidant effects alone. In this review, I focused on the bioavailability of phytobiotics and the possible mechanisms underlying their overall effects on intestinal barrier functions, inflammatory status, gut microbiota, systemic inflammation, and metabolism, rather than the specific effects of each compound. I also discuss the possible mechanisms by which phytobiotics contribute to growth promotion in chickens.
Collapse
Affiliation(s)
- Motoi Kikusato
- Animal Nutrition, Life Sciences, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan
| |
Collapse
|
32
|
Desantis S, Galosi L, Santamaria N, Roncarati A, Biagini L, Rossi G. Modulation of Morphology and Glycan Composition of Mucins in Farmed Guinea Fowl ( Numida meleagris) Intestine by the Multi-Strain Probiotic Slab51 ®. Animals (Basel) 2021; 11:495. [PMID: 33668637 PMCID: PMC7918860 DOI: 10.3390/ani11020495] [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: 12/30/2020] [Revised: 02/07/2021] [Accepted: 02/09/2021] [Indexed: 11/24/2022] Open
Abstract
Probiotics have become highly recognized as supplements for poultry.Since gut health can be considered synonymous withanimal health, the effects of probiotic Slab51® on the morphology and the glycan composition of guineafowlintestine were examined. The probiotics were added in drinking water (2 × 1011 UFC/L) throughout the grow-out cycle.Birds were individually weighed andslaughtered after four months. Samples from the duodenum, ileum and caecum were collected and processed for morphological, morphometric, conventional and lectin glycohistochemical studies.The results were analyzed for statistical significance by Student's t test. Compared with control samples, probiotic group revealed (1) significant increase in villus height (p < 0.001 in duodenum and ileum; p < 0.05 in caecum), crypt depth (p < 0.001 in duodenum and caecum; p < 0.05 in ileum) and goblet cells (GCs) per villus (p < 0.001) in all investigated tracts; (2) increase in galactoseβl,3N-acetylgalacyosamine(Galβl,3GalNAc)terminating O-glycans and αl,2-fucosylated glycans secretory GCs in the duodenum; (3) increase in α2,6-sialoglycans and high-mannose N-linked glycans secretory GCs but reduction in GCs-secreting sulfoglycans in the ileum; (4) increase in Galβl,3GalNAc and high-mannose N-linked glycans secretory GCs and decrease in GCs-producing sulfomucins in the caecum; (5) increase in the numbers of crypt cells containing sulfate and non-sulfated acidic glycans. Overall, dietary Slab51® induces morphological and region-specific changes in glycoprotein composition of guinea fowl intestine, promoting gut health.
Collapse
Affiliation(s)
- Salvatore Desantis
- Department of Emergency and Organ Transplantation (DETO), University of Bari Aldo Moro, S.P. 62 per Casamassima Km 3, 70010 Valenzano (Bari), Italy;
| | - Livio Galosi
- School of Biosciences and Veterinary medicine, University of Camerino, Via Circonvallazione 93/95, 62024 Matelica (M.C.), Italy; (A.R.); (L.B.); (G.R.)
| | - Nicoletta Santamaria
- Department of Emergency and Organ Transplantation (DETO), University of Bari Aldo Moro, S.P. 62 per Casamassima Km 3, 70010 Valenzano (Bari), Italy;
| | - Alessandra Roncarati
- School of Biosciences and Veterinary medicine, University of Camerino, Via Circonvallazione 93/95, 62024 Matelica (M.C.), Italy; (A.R.); (L.B.); (G.R.)
| | - Lucia Biagini
- School of Biosciences and Veterinary medicine, University of Camerino, Via Circonvallazione 93/95, 62024 Matelica (M.C.), Italy; (A.R.); (L.B.); (G.R.)
| | - Giacomo Rossi
- School of Biosciences and Veterinary medicine, University of Camerino, Via Circonvallazione 93/95, 62024 Matelica (M.C.), Italy; (A.R.); (L.B.); (G.R.)
| |
Collapse
|
33
|
Benktander J, Sundh H, Sundell K, Murugan AVM, Venkatakrishnan V, Padra JT, Kolarevic J, Terjesen BF, Gorissen M, Lindén SK. Stress Impairs Skin Barrier Function and Induces α2-3 Linked N-Acetylneuraminic Acid and Core 1 O-Glycans on Skin Mucins in Atlantic Salmon, Salmo salar. Int J Mol Sci 2021; 22:ijms22031488. [PMID: 33540792 PMCID: PMC7867331 DOI: 10.3390/ijms22031488] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Revised: 01/26/2021] [Accepted: 01/27/2021] [Indexed: 02/07/2023] Open
Abstract
The skin barrier consists of mucus, primarily comprising highly glycosylated mucins, and the epithelium. Host mucin glycosylation governs interactions with pathogens and stress is associated with impaired epithelial barrier function. We characterized Atlantic salmon skin barrier function during chronic stress (high density) and mucin O-glycosylation changes in response to acute and chronic stress. Fish held at low (LD: 14–30 kg/m3) and high densities (HD: 50-80 kg/m3) were subjected to acute stress 24 h before sampling at 17 and 21 weeks after start of the experiment. Blood parameters indicated primary and secondary stress responses at both sampling points. At the second sampling, skin barrier function towards molecules was reduced in the HD compared to the LD group (Papp mannitol; p < 0.01). Liquid chromatography–mass spectrometry revealed 81 O-glycan structures from the skin. Fish subjected to both chronic and acute stress had an increased proportion of large O-glycan structures. Overall, four of the O-glycan changes have potential as indicators of stress, especially for the combined chronic and acute stress. Stress thus impairs skin barrier function and induces glycosylation changes, which have potential to both affect interactions with pathogens and serve as stress indicators.
Collapse
Affiliation(s)
- John Benktander
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Box 440, Medicinaregatan 9C, 405 30 Gothenburg, Sweden; (J.B.); (A.V.M.M.); (V.V.); (J.T.P.)
| | - Henrik Sundh
- Department of Biological and Environmental Sciences, University of Gothenburg, 405 30 Gothenburg, Sweden; (H.S.); (K.S.)
| | - Kristina Sundell
- Department of Biological and Environmental Sciences, University of Gothenburg, 405 30 Gothenburg, Sweden; (H.S.); (K.S.)
| | - Abarna V. M. Murugan
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Box 440, Medicinaregatan 9C, 405 30 Gothenburg, Sweden; (J.B.); (A.V.M.M.); (V.V.); (J.T.P.)
| | - Vignesh Venkatakrishnan
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Box 440, Medicinaregatan 9C, 405 30 Gothenburg, Sweden; (J.B.); (A.V.M.M.); (V.V.); (J.T.P.)
| | - János Tamás Padra
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Box 440, Medicinaregatan 9C, 405 30 Gothenburg, Sweden; (J.B.); (A.V.M.M.); (V.V.); (J.T.P.)
| | | | | | - Marnix Gorissen
- Radboud University, Institute for Water and Wetland Research, Department of Animal Ecology & Physiology, Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands;
| | - Sara K. Lindén
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Box 440, Medicinaregatan 9C, 405 30 Gothenburg, Sweden; (J.B.); (A.V.M.M.); (V.V.); (J.T.P.)
- Correspondence: ; Tel.: +46-(0)-31-786-3057
| |
Collapse
|
34
|
Gill Mucus and Gill Mucin O-glycosylation in Healthy and Amebic Gill Disease-Affected Atlantic Salmon. Microorganisms 2020; 8:microorganisms8121871. [PMID: 33256221 PMCID: PMC7768351 DOI: 10.3390/microorganisms8121871] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 11/23/2020] [Accepted: 11/24/2020] [Indexed: 01/17/2023] Open
Abstract
Amoebic gill disease (AGD) causes poor performance and death in salmonids. Mucins are mainly comprised by carbohydrates and are main components of the mucus covering the gill. Since glycans regulate pathogen binding and growth, glycosylation changes may affect susceptibility to primary and secondary infections. We investigated gill mucin O-glycosylation from Atlantic salmon with and without AGD using liquid chromatography–mass spectrometry. Gill mucin glycans were larger and more complex, diverse and fucosylated than skin mucins. Confocal microscopy revealed that fucosylated mucus coated sialylated mucus strands in ex vivo gill mucus. Terminal HexNAcs were more abundant among O-glycans from AGD-affected Atlantic salmon, whereas core 1 structures and structures with acidic moieties such as N-acetylneuraminic acid (NeuAc) and sulfate groups were less abundant compared to non-infected fish. The fucosylated and NeuAc-containing O-glycans were inversely proportional, with infected fish on the lower scale of NeuAc abundance and high on fucosylated structures. The fucosylated epitopes were of three types: Fuc-HexNAc-R, Gal-[Fuc-]HexNAc-R and HexNAc-[Fuc-]HexNAc-R. These blood group-like structures could be an avenue to diversify the glycan repertoire to limit infection in the exposed gills. Furthermore, care must be taken when using skin mucus as proxy for gill mucus, as gill mucins are distinctly different from skin mucins.
Collapse
|
35
|
González-Morelo KJ, Vega-Sagardía M, Garrido D. Molecular Insights Into O-Linked Glycan Utilization by Gut Microbes. Front Microbiol 2020; 11:591568. [PMID: 33224127 PMCID: PMC7674204 DOI: 10.3389/fmicb.2020.591568] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 10/20/2020] [Indexed: 12/16/2022] Open
Abstract
O-linked glycosylation is a post-translational modification found mainly in eukaryotic cells, which covalently attaches oligosaccharides to secreted proteins in certain threonine or serine residues. Most of O-glycans have N-acetylgalactosamine (GalNAc) as a common core. Several glycoproteins, such as mucins (MUCs), immunoglobulins, and caseins are examples of O-glycosylated structures. These glycans are further elongated with other monosaccharides and sulfate groups. Some of them could be found in dairy foods, while others are produced endogenously, in both cases interacting with the gut microbiota. Interestingly, certain gut microbes can access, release, and consume O-linked glycans as a carbon source. Among these, Akkermansia muciniphila, Bifidobacterium bifidum, and Bacteroides thetaiotaomicron are prominent O-linked glycan utilizers. Their consumption strategies include specialized α-fucosidases and α-sialidases, in addition to endo-α-N-acetylgalactosaminidases that release galacto-N-biose (GNB) from peptides backbones. O-linked glycan utilization by certain gut microbes represents an important niche that allows them to predominate and modulate host responses such as inflammation. Here, we focus on the distinct molecular mechanisms of consumption of O-linked GalNAc glycans by prominent gut microbes, especially from mucin and casein glycomacropeptide (GMP), highlighting the potential of these structures as emerging prebiotics.
Collapse
Affiliation(s)
| | | | - Daniel Garrido
- Department of Chemical and Bioprocess Engineering, School of Engineering, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
36
|
Firmino JP, Vallejos-Vidal E, Sarasquete C, Ortiz-Delgado JB, Balasch JC, Tort L, Estevez A, Reyes-López FE, Gisbert E. Unveiling the effect of dietary essential oils supplementation in Sparus aurata gills and its efficiency against the infestation by Sparicotyle chrysophrii. Sci Rep 2020; 10:17764. [PMID: 33082387 PMCID: PMC7576129 DOI: 10.1038/s41598-020-74625-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 09/29/2020] [Indexed: 12/11/2022] Open
Abstract
A microencapsulated feed additive composed by garlic, carvacrol and thymol essential oils (EOs) was evaluated regarding its protective effect in gills parasitized by Sparicotyle chrysophrii in Sparus aurata. A nutritional trial (65 days) followed by a cohabitation challenge with parasitized fish (39 days) were performed. Transcriptomic analysis by microarrays of gills of fish fed the EOs diet showed an up-regulation of genes related to biogenesis, vesicular transport and exocytosis, leukocyte-mediated immunity, oxidation–reduction and overall metabolism processes. The functional network obtained indicates a tissue-specific pro-inflammatory immune response arbitrated by degranulating acidophilic granulocytes, sustained by antioxidant and anti-inflammatory responses. The histochemical study of gills also showed an increase of carboxylate glycoproteins containing sialic acid in mucous and epithelial cells of fish fed the EOs diet, suggesting a mucosal defence mechanism through the modulation of mucin secretions. The outcomes of the in vivo challenge supported the transcriptomic results obtained from the nutritional trial, where a significant reduction of 78% in the abundance of S. chrysophrii total parasitation and a decrease in the prevalence of most parasitic developmental stages evaluated were observed in fish fed the EOs diet. These results suggest that the microencapsulation of garlic, carvacrol and thymol EOs could be considered an effective natural dietary strategy with antiparasitic properties against the ectoparasite S. chrysophrii.
Collapse
Affiliation(s)
- Joana P Firmino
- IRTA, Centre de Sant Carles de la Ràpita (IRTA-SCR), Aquaculture Program, Crta. Poble Nou km 5.5, 43540, Sant Carles de la Ràpita, Spain.,TECNOVIT-FARMFAES, S.L. Pol. Ind. Les Sorts, parc. 10, 43365, Alforja, Spain.,PhD Program in Aquaculture, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain
| | - Eva Vallejos-Vidal
- Centro de Biotecnología Acuícola, Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
| | - Carmen Sarasquete
- Instituto de Ciencias Marinas de Andalucía (ICMAN-CSIC), Universidad de Cádiz, Campus Universitario Río San Pedro, Puerto Real, Cádiz, Spain
| | - Juan B Ortiz-Delgado
- Instituto de Ciencias Marinas de Andalucía (ICMAN-CSIC), Universidad de Cádiz, Campus Universitario Río San Pedro, Puerto Real, Cádiz, Spain
| | - Joan Carles Balasch
- Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain
| | - Lluis Tort
- Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain
| | - Alicia Estevez
- IRTA, Centre de Sant Carles de la Ràpita (IRTA-SCR), Aquaculture Program, Crta. Poble Nou km 5.5, 43540, Sant Carles de la Ràpita, Spain
| | - Felipe E Reyes-López
- Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain.
| | - Enric Gisbert
- IRTA, Centre de Sant Carles de la Ràpita (IRTA-SCR), Aquaculture Program, Crta. Poble Nou km 5.5, 43540, Sant Carles de la Ràpita, Spain.
| |
Collapse
|
37
|
Rastad A. Effects of antibiotic replacement with garlic powder and probiotic on performance, carcass characteristics, oxidative enzymes and intestinal morphology of broiler chickens. ACTA SCIENTIARUM: ANIMAL SCIENCES 2020. [DOI: 10.4025/actascianimsci.v42i1.48734] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
An experiment was conducted to evaluate the effects of probiotic and garlic powder instead of antibiotic on performance, carcass characteristics, oxidative enzymes and intestinal morphology of broiler chickens. A total of 200 one-day-old male broiler chicks were used in a completely randomized design. The experimental groups were including control group (without any additives) or CG, antibiotic group or AG, garlic powder group or GG, probiotic group or PG and garlic powder plus probiotic group or GPG. The broilers were weighted at the end of days 10, 24 and 42 to evaluate the body performance. At the end of experiment, four broilers randomly selected from each replicate to blood sampling and carcass traits measurement (2 chickens for each one). The use of GG significantly decreased feed intake than AG (p < 0.05) which approved in GPG when probiotic added to GG (p < 0.05). All groups, exception GG showed less feed intake than CG between days 0 to 42 of experiment (p < 0.05). GPG shows significant differences than CG between days 21 to 42 and 0 to 42 and also than GG between days 0 to 42. The use of GG and GPG significantly increased liver enzyme activities (p < 0.05). AG, PG and GPG showed a higher height and width villi than CG. So simultaneous use of probiotic and garlic powder can be a suitable alternative to antibiotics to normal performance and liver function
Collapse
|
38
|
Auffret MD, Stewart RD, Dewhurst RJ, Duthie CA, Watson M, Roehe R. Identification of Microbial Genetic Capacities and Potential Mechanisms Within the Rumen Microbiome Explaining Differences in Beef Cattle Feed Efficiency. Front Microbiol 2020; 11:1229. [PMID: 32582125 PMCID: PMC7292206 DOI: 10.3389/fmicb.2020.01229] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 05/14/2020] [Indexed: 12/15/2022] Open
Abstract
In this study, Bos Taurus cattle offered one high concentrate diet (92% concentrate-8% straw) during two independent trials allowed us to classify 72 animals comprising of two cattle breeds as "Low" or "High" feed efficiency groups. Digesta samples were taken from individual beef cattle at the abattoir. After metagenomic sequencing, the rumen microbiome composition and genes were determined. Applying a targeted approach based on current biological evidence, 27 genes associated with host-microbiome interaction activities were selected. Partial least square analysis enabled the identification of the most significant genes and genera of feed efficiency (VIP > 0.8) across years of the trial and breeds when comparing all potential genes or genera together. As a result, limited number of genes explained about 40% of the variability in both feed efficiency indicators. Combining information from rumen metagenome-assembled genomes and partial least square analysis results, microbial genera carrying these genes were determined and indicated that a limited number of important genera impacting on feed efficiency. In addition, potential mechanisms explaining significant difference between Low and High feed efficiency animals were analyzed considering, based on the literature, their gastrointestinal location of action. High feed efficiency animals were associated with microbial species including several Eubacterium having the genetic capacity to form biofilm or releasing metabolites like butyrate or propionate known to provide a greater contribution to cattle energy requirements compared to acetate. Populations associated with fucose sensing or hemolysin production, both mechanisms specifically described in the lower gut by activating the immune system to compete with pathogenic colonizers, were also identified to affect feed efficiency using rumen microbiome information. Microbial mechanisms associated with low feed efficiency animals involved potential pathogens within Proteobacteria and Spirochaetales, releasing less energetic substrates (e.g., acetate) or producing sialic acid to avoid the host immune system. Therefore, this study focusing on genes known to be involved in host-microbiome interaction improved the identification of rumen microbial genetic capacities and potential mechanisms significantly impacting on feed efficiency in beef cattle fed high concentrate diet.
Collapse
Affiliation(s)
| | - Robert D. Stewart
- Division of Genetics and Genomics, The Roslin Institute and R(D)SVS, University of Edinburgh, Edinburgh, United Kingdom
| | | | | | - Mick Watson
- Division of Genetics and Genomics, The Roslin Institute and R(D)SVS, University of Edinburgh, Edinburgh, United Kingdom
- Edinburgh Genomics, The Roslin Institute and R(D)SVS, University of Edinburgh, Edinburgh, United Kingdom
| | - Rainer Roehe
- Scotland’s Rural College (SRUC), Edinburgh, United Kingdom
| |
Collapse
|
39
|
Zhu L, Lu X, Liu L, Voglmeir J, Zhong X, Yu Q. Akkermansia muciniphila protects intestinal mucosa from damage caused by S. pullorum by initiating proliferation of intestinal epithelium. Vet Res 2020; 51:34. [PMID: 32138776 PMCID: PMC7057645 DOI: 10.1186/s13567-020-00755-3] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 01/15/2020] [Indexed: 12/16/2022] Open
Abstract
Akkermansia muciniphila, a novel mucin-degrading bacterium, has been demonstrated to prevent the development of obesity and related complications. However, whether it can protect poultry from intestinal mucosal damage by enteropathogens has never been mentioned. In this study, we found that A. muciniphila colonized in the intestine and then relieved intestinal mucosal damage in chicks caused by S. pullorum, including anatomical and morphological damage, alleviation of body weight and intestinal inflammation. The repair process activated by A. muciniphila is accompanied by an increase in the number of goblet cells in the chick’s intestine and an up-regulation of Mucin 2 and trefoil factor 2 (Tff2). In addition, we also demonstrate that A. muciniphila improved colon length, crypt depth, increased the proliferating cell nuclear antigen, with the accelerated proliferation of intestinal epithelium through Wnt/β-catenin signaling pathway, thereby restoring the damaged intestinal mucosa. This study suggests that A. muciniphila activates the proliferation of intestinal cells protecting the intestinal barrier, thus relieving infection with S. pullorum in chickens.
Collapse
Affiliation(s)
- Linda Zhu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Weigang 1, Nanjing, 210095, Jiangsu, China
| | - Xiaoxi Lu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Weigang 1, Nanjing, 210095, Jiangsu, China
| | - Li Liu
- Glycomics and Glycan Bioengineering Research Center (GGBRC), College of Food Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Josef Voglmeir
- Glycomics and Glycan Bioengineering Research Center (GGBRC), College of Food Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Xiang Zhong
- College of Animal Science and Technology, Nanjing Agricultural University, Weigang 1, Nanjing, 210095, Jiangsu, China.
| | - Qinghua Yu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Weigang 1, Nanjing, 210095, Jiangsu, China.
| |
Collapse
|
40
|
Padra JT, Pagneux Q, Bouckaert J, Jijie R, Sundh H, Boukherroub R, Szunerits S, Lindén SK. Mucin modified SPR interfaces for studying the effect of flow on pathogen binding to Atlantic salmon mucins. Biosens Bioelectron 2019; 146:111736. [PMID: 31586762 DOI: 10.1016/j.bios.2019.111736] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 09/26/2019] [Accepted: 09/27/2019] [Indexed: 01/10/2023]
Abstract
Knowledge on host-pathogen interactions contributes to the development of approaches to alleviate infectious disease. In this work, we developed a surface plasmon resonance (SPR) based method for investigating bacteria/mucins interactions. Furthermore, we investigated adhesion of three pathogens, Aeromonas salmonicida, Aeromonas hydrophila and Vibrio harveyi, to Atlantic salmon mucins isolated from different epithelial sites, using SPR and microtiter-based binding assays. We demonstrated that performing bacterial binding assays to mucins using SPR is feasible and has advantages over microtiter-based binding assays, especially under flow conditions. The fluid flow in the SPR is linear and continuous and SPR enables real-time reading of mucin-bacterial bonds, which provides an in vivo-like setup for analysis of bacterial binding to mucins. The variation between technical replicates was smaller using SPR detection compared to the adenosine 5'-triphosphate (ATP) bioluminescence assay in microtiter plates. Furthermore, we demonstrated that the effect of flow on pathogen-mucin interaction is significant and that bacterial adhesion differ non-linearly with flow rates and depend on the epithelial source of the mucin.
Collapse
Affiliation(s)
- János Tamás Padra
- Department of Medical Chemistry and Cell Biology, University of Gothenburg, Gothenburg, SE-405 30, Sweden
| | - Quentin Pagneux
- Univ. Lille, CNRS, Centrale Lille, ISEN, Univ. Valenciennes, UMR 8520, IEMN, F-59000, Lille, France
| | - Julie Bouckaert
- Unité de Glycobiologie Structurale et Fonctionnelle (UGSF), Univ. Lille, CNRS, UMR 8576, 59658, Villeneuve d'Ascq, France
| | - Roxana Jijie
- Univ. Lille, CNRS, Centrale Lille, ISEN, Univ. Valenciennes, UMR 8520, IEMN, F-59000, Lille, France
| | - Henrik Sundh
- Department of Biological and Environmental Sciences, University of Gothenburg, Gothenburg, Sweden
| | - Rabah Boukherroub
- Univ. Lille, CNRS, Centrale Lille, ISEN, Univ. Valenciennes, UMR 8520, IEMN, F-59000, Lille, France
| | - Sabine Szunerits
- Univ. Lille, CNRS, Centrale Lille, ISEN, Univ. Valenciennes, UMR 8520, IEMN, F-59000, Lille, France.
| | - Sara K Lindén
- Department of Medical Chemistry and Cell Biology, University of Gothenburg, Gothenburg, SE-405 30, Sweden.
| |
Collapse
|
41
|
Jiang S, El-Senousey HK, Fan Q, Lin X, Gou Z, Li L, Wang Y, Fouad AM, Jiang Z. Effects of dietary threonine supplementation on productivity and expression of genes related to protein deposition and amino acid transportation in breeder hens of yellow-feathered chicken and their offspring. Poult Sci 2019; 98:6826-6836. [PMID: 31504946 PMCID: PMC6870553 DOI: 10.3382/ps/pez420] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 08/30/2019] [Indexed: 01/07/2023] Open
Abstract
This study investigated the effects of the dietary threonine (Thr) levels on the performance, offspring traits, embryo amino acid transportation, and protein deposition in breeder hens of yellow-feathered chickens. In total, 720 breeder hens of Lingnan yellow-feathered chickens were randomly assigned to 1 of 6 dietary treatments, with 6 replicates per treatment (20 birds per replicate). The breeder hens were fed either basal diet (Thr = 0.38%) or basal diet supplemented with 0.12, 0.24, 0.36, 0.48, or 0.60% Thr from 197 to 266 D. There was a positive response in terms of the laying rate after adding different levels of Thr to the diet, but no significant effects on the average daily gain, average daily egg weight, feed conversion ratio, average broken eggs, and unqualified egg rate (P > 0.05). However, the eggshell strength and eggshell percentage decreased in a linear manner as the dietary Thr concentration increased (P = 0.05). Dietary supplementation with Thr had significant effects on the expression of mucin 2 (MUC2) in the uterus and zonula occludens protein 1 (ZO-1) in the duodenum of breeders (P < 0.05). In chick embryos at embryonic age 18 D, significant upregulation of poultry target of rapamycin (pTOR) occurred in the liver and breast muscle, as well as threonine dehydrogenase (TDH) in the thigh, and aminopeptidase (ANPEP) (P < 0.05) in the duodenum and ileum due to dietary Thr supplementation, but there were no effects on MUC2 expression in the duodenum and ileum (P > 0.05). The livability of the progeny broilers tended to increase with the dietary Thr concentration (quadratic, P = 0.08). Thus, dietary supplementation with Thr had positive effects on the laying production by breeder hens and offspring performance, and it also regulated the expression levels of genes related to amino acid transportation and protein deposition. The optimal dietary Thr concentration that maximized the laying rate in yellow-feathered chicken breeders aged 197 to 266 D was 0.68% according to quadratic regression analysis.
Collapse
Affiliation(s)
- Shouqun Jiang
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, State Key Laboratory of Livestock and Poultry Breeding, Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture, Guangdong Key Laboratory of Animal Breeding and Nutrition, Guangdong Public Laboratory of Animal Breeding and Nutrition, 510640 Guangzhou, P. R. China,Corresponding author:
| | - HebatAllah Kasem El-Senousey
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, State Key Laboratory of Livestock and Poultry Breeding, Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture, Guangdong Key Laboratory of Animal Breeding and Nutrition, Guangdong Public Laboratory of Animal Breeding and Nutrition, 510640 Guangzhou, P. R. China,Department of Animal Production, Faculty of Agriculture, Cairo University, Giza 12613, Egypt
| | - Qiuli Fan
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, State Key Laboratory of Livestock and Poultry Breeding, Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture, Guangdong Key Laboratory of Animal Breeding and Nutrition, Guangdong Public Laboratory of Animal Breeding and Nutrition, 510640 Guangzhou, P. R. China
| | - Xiajing Lin
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, State Key Laboratory of Livestock and Poultry Breeding, Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture, Guangdong Key Laboratory of Animal Breeding and Nutrition, Guangdong Public Laboratory of Animal Breeding and Nutrition, 510640 Guangzhou, P. R. China
| | - Zhongyong Gou
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, State Key Laboratory of Livestock and Poultry Breeding, Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture, Guangdong Key Laboratory of Animal Breeding and Nutrition, Guangdong Public Laboratory of Animal Breeding and Nutrition, 510640 Guangzhou, P. R. China
| | - Long Li
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, State Key Laboratory of Livestock and Poultry Breeding, Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture, Guangdong Key Laboratory of Animal Breeding and Nutrition, Guangdong Public Laboratory of Animal Breeding and Nutrition, 510640 Guangzhou, P. R. China
| | - Yibing Wang
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, State Key Laboratory of Livestock and Poultry Breeding, Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture, Guangdong Key Laboratory of Animal Breeding and Nutrition, Guangdong Public Laboratory of Animal Breeding and Nutrition, 510640 Guangzhou, P. R. China
| | - Ahmed Mohamed Fouad
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, State Key Laboratory of Livestock and Poultry Breeding, Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture, Guangdong Key Laboratory of Animal Breeding and Nutrition, Guangdong Public Laboratory of Animal Breeding and Nutrition, 510640 Guangzhou, P. R. China,Department of Animal Production, Faculty of Agriculture, Cairo University, Giza 12613, Egypt
| | - Zongyong Jiang
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, State Key Laboratory of Livestock and Poultry Breeding, Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture, Guangdong Key Laboratory of Animal Breeding and Nutrition, Guangdong Public Laboratory of Animal Breeding and Nutrition, 510640 Guangzhou, P. R. China,Corresponding author:
| |
Collapse
|
42
|
Näpflin K, O’Connor EA, Becks L, Bensch S, Ellis VA, Hafer-Hahmann N, Harding KC, Lindén SK, Olsen MT, Roved J, Sackton TB, Shultz AJ, Venkatakrishnan V, Videvall E, Westerdahl H, Winternitz JC, Edwards SV. Genomics of host-pathogen interactions: challenges and opportunities across ecological and spatiotemporal scales. PeerJ 2019; 7:e8013. [PMID: 31720122 PMCID: PMC6839515 DOI: 10.7717/peerj.8013] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 10/08/2019] [Indexed: 12/13/2022] Open
Abstract
Evolutionary genomics has recently entered a new era in the study of host-pathogen interactions. A variety of novel genomic techniques has transformed the identification, detection and classification of both hosts and pathogens, allowing a greater resolution that helps decipher their underlying dynamics and provides novel insights into their environmental context. Nevertheless, many challenges to a general understanding of host-pathogen interactions remain, in particular in the synthesis and integration of concepts and findings across a variety of systems and different spatiotemporal and ecological scales. In this perspective we aim to highlight some of the commonalities and complexities across diverse studies of host-pathogen interactions, with a focus on ecological, spatiotemporal variation, and the choice of genomic methods used. We performed a quantitative review of recent literature to investigate links, patterns and potential tradeoffs between the complexity of genomic, ecological and spatiotemporal scales undertaken in individual host-pathogen studies. We found that the majority of studies used whole genome resolution to address their research objectives across a broad range of ecological scales, especially when focusing on the pathogen side of the interaction. Nevertheless, genomic studies conducted in a complex spatiotemporal context are currently rare in the literature. Because processes of host-pathogen interactions can be understood at multiple scales, from molecular-, cellular-, and physiological-scales to the levels of populations and ecosystems, we conclude that a major obstacle for synthesis across diverse host-pathogen systems is that data are collected on widely diverging scales with different degrees of resolution. This disparity not only hampers effective infrastructural organization of the data but also data granularity and accessibility. Comprehensive metadata deposited in association with genomic data in easily accessible databases will allow greater inference across systems in the future, especially when combined with open data standards and practices. The standardization and comparability of such data will facilitate early detection of emerging infectious diseases as well as studies of the impact of anthropogenic stressors, such as climate change, on disease dynamics in humans and wildlife.
Collapse
Affiliation(s)
- Kathrin Näpflin
- Department of Organismic and Evolutionary Biology and Museum of Comparative Zoology, Harvard University, Cambridge, MA, United States of America
| | - Emily A. O’Connor
- Molecular Ecology and Evolution Lab, Department of Biology, Lund University, Lund, Sweden
| | - Lutz Becks
- Aquatic Ecology and Evolution, Limnological Institute University Konstanz, Konstanz, Germany
| | - Staffan Bensch
- Molecular Ecology and Evolution Lab, Department of Biology, Lund University, Lund, Sweden
| | - Vincenzo A. Ellis
- Molecular Ecology and Evolution Lab, Department of Biology, Lund University, Lund, Sweden
| | - Nina Hafer-Hahmann
- Department of Evolutionary Ecology, Max Planck Institute for Evolutionary Biology, Plön, Germany
- EAWAG, Swiss Federal Institute of Aquatic Science and Technology, Dübendorf, Switzerland
| | - Karin C. Harding
- Department of Biological and Environmental Sciences, University of Gothenburg, Gothenburg, Sweden
- Gothenburg Centre for Advanced Studies in Science and Technology, Chalmers University of Technology and University of Gothenburg, Gothenburg, Sweden
| | - Sara K. Lindén
- Department of Medical Chemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Morten T. Olsen
- Section for Evolutionary Genomics, Natural History Museum of Denmark, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Jacob Roved
- Molecular Ecology and Evolution Lab, Department of Biology, Lund University, Lund, Sweden
| | - Timothy B. Sackton
- Informatics Group, Harvard University, Cambridge, MA, United States of America
| | - Allison J. Shultz
- Ornithology Department, Natural History Museum of Los Angeles County, Los Angeles, CA, United States of America
| | - Vignesh Venkatakrishnan
- Department of Medical Chemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Elin Videvall
- Molecular Ecology and Evolution Lab, Department of Biology, Lund University, Lund, Sweden
- Center for Conservation Genomics, Smithsonian Conservation Biology Institute, National Zoological Park, Washington, DC, United States of America
| | - Helena Westerdahl
- Molecular Ecology and Evolution Lab, Department of Biology, Lund University, Lund, Sweden
| | - Jamie C. Winternitz
- Department of Evolutionary Ecology, Max Planck Institute for Evolutionary Biology, Plön, Germany
- Department of Animal Behaviour, Bielefeld University, Bielefeld, Germany
| | - Scott V. Edwards
- Department of Organismic and Evolutionary Biology and Museum of Comparative Zoology, Harvard University, Cambridge, MA, United States of America
- Gothenburg Centre for Advanced Studies in Science and Technology, Chalmers University of Technology and University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
43
|
Role of Sialic Acid in Brachyspira hyodysenteriae Adhesion to Pig Colonic Mucins. Infect Immun 2019; 87:IAI.00889-18. [PMID: 30988055 DOI: 10.1128/iai.00889-18] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Accepted: 04/05/2019] [Indexed: 12/21/2022] Open
Abstract
Infection with Brachyspira hyodysenteriae results in mucoid hemorrhagic diarrhea. This pathogen is associated with the colonic mucus layer, mainly composed of mucins. Infection regulates mucin O-glycosylation in the colon and increases mucin secretion as well as B. hyodysenteriae binding sites on mucins. Here, we analyzed potential mucin epitopes for B. hyodysenteriae adhesion in the colon, as well as the effect of colonic mucins on bacterial growth. Associations between B. hyodysenteriae binding to pig colonic mucins and mucin glycan data showed that B. hyodysenteriae binding was associated with the presence of N-glycolylneuraminic acid (NeuGc) on mucins. The role of sialic acid in B. hyodysenteriae adhesion was analyzed after the removal of sialic acid residues on the mucins by enzymatic treatment with sialidase A, which decreased bacterial binding to the mucins. The effect of pig colonic mucins on B. hyodysenteriae growth was determined in carbohydrate-free medium. B. hyodysenteriae growth increased in the presence of mucins from two out of five infected pigs, suggesting utilization of mucins as a carbon source for growth. Additionally, bacterial growth was enhanced by free sialic acid and N-acetylglucosamine. The results highlight a role of sialic acid as an adhesion epitope for B. hyodysenteriae interaction with colonic mucins. Furthermore, the mucin response and glycosylation changes exerted in the colon during B. hyodysenteriae infection result in a potentially favorable environment for pathogen growth in the intestinal mucus layer.
Collapse
|
44
|
Helicobacter suis infection alters glycosylation and decreases the pathogen growth inhibiting effect and binding avidity of gastric mucins. Mucosal Immunol 2019; 12:784-794. [PMID: 30846831 DOI: 10.1038/s41385-019-0154-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 01/11/2019] [Accepted: 02/10/2019] [Indexed: 02/07/2023]
Abstract
Helicobacter suis is the most prevalent non-Helicobacter pylori Helicobacter species in the human stomach and is associated with chronic gastritis, peptic ulcer disease, and gastric mucosa-associated lymphoid tissue (MALT) lymphoma. H. suis colonizes the gastric mucosa of 60-95% of pigs at slaughter age, and is associated with chronic gastritis, decreased weight gain, and ulcers. Here, we show that experimental H. suis infection changes the mucin composition and glycosylation, decreasing the amount of H. suis-binding glycan structures in the pig gastric mucus niche. Similarly, the H. suis-binding ability of mucins from H. pylori-infected humans is lower than that of noninfected individuals. Furthermore, the H. suis growth-inhibiting effect of mucins from both noninfected humans and pigs is replaced by a growth-enhancing effect by mucins from infected individuals/pigs. Thus, Helicobacter spp. infections impair the mucus barrier by decreasing the H. suis-binding ability of the mucins and by decreasing the antiprolific activity that mucins can have on H. suis. Inhibition of these mucus-based defenses creates a more stable and inhabitable niche for H. suis. This is likely of importance for long-term colonization and outcome of infection, and reversing these impairments may have therapeutic benefits.
Collapse
|
45
|
Venkatakrishnan V, Padra JT, Sundh H, Sundell K, Jin C, Langeland M, Carlberg H, Vidakovic A, Lundh T, Karlsson NG, Lindén SK. Exploring the Arctic Charr Intestinal Glycome: Evidence of Increased N-Glycolylneuraminic Acid Levels and Changed Host-Pathogen Interactions in Response to Inflammation. J Proteome Res 2019; 18:1760-1773. [PMID: 30848132 DOI: 10.1021/acs.jproteome.8b00973] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Disease outbreaks are a limiting factor for the sustainable development of the aquaculture industry. The intestinal tract is covered by a mucus layer mainly comprised by highly glycosylated proteins called mucins. Mucins regulate pathogen adhesion, growth, and virulence, and the glycans are vital for these functions. We analyzed intestinal mucin O-glycans on mucins from control and full-fat extruded soy-bean-fed (known to cause enteritis) Arctic charr using liquid chromatography-tandem mass spectrometry. In total, 56 glycans were identified on Arctic charr intestinal mucins, with a high prevalence of core-5-type and sialylated O-glycans. Disialic-acid-epitope-containing structures including NeuAcα2,8NeuAc, NeuAc(Gc)α2,8NeuGc(Ac), and NeuGcα2,8NeuGc were the hallmark of Arctic charr intestinal mucin glycosylation. Arctic charr fed with soy bean meal diet had lower (i) number of structures detected, (ii) interindividual variation, and (iii) N-glycolylneuraminic-acid-containing glycans compared with control Arctic charr. Furthermore, Aeromonas salmonicida grew less in response to mucins from inflamed Arctic charr than from the control group. The Arctic charr glycan repertoire differed from that of Atlantic salmon. In conclusion, the loss of N-glycolylneuraminic acid may be a biomarker for inflammation in Arctic char, and inflammation-induced glycosylation changes affect host-pathogen interactions.
Collapse
Affiliation(s)
- Vignesh Venkatakrishnan
- Department of Medical Chemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy , University of Gothenburg , Gothenburg 40530 , Sweden
| | - János T Padra
- Department of Medical Chemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy , University of Gothenburg , Gothenburg 40530 , Sweden
| | - Henrik Sundh
- Department of Biological and Environmental Sciences , University of Gothenburg , Gothenburg 40530 , Sweden
| | - Kristina Sundell
- Department of Biological and Environmental Sciences , University of Gothenburg , Gothenburg 40530 , Sweden
| | - Chunsheng Jin
- Department of Medical Chemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy , University of Gothenburg , Gothenburg 40530 , Sweden
| | - Markus Langeland
- Department of Animal Nutrition and Management , Swedish University of Agricultural Science , Uppsala 75007 , Sweden
| | - Hanna Carlberg
- Department of Wildlife, Fish, and Environmental Studies , Swedish University of Agricultural Science , Umeå 90736 , Sweden
| | - Aleksander Vidakovic
- Department of Animal Nutrition and Management , Swedish University of Agricultural Science , Uppsala 75007 , Sweden
| | - Torbjörn Lundh
- Department of Animal Nutrition and Management , Swedish University of Agricultural Science , Uppsala 75007 , Sweden
| | - Niclas G Karlsson
- Department of Medical Chemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy , University of Gothenburg , Gothenburg 40530 , Sweden
| | - Sara K Lindén
- Department of Medical Chemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy , University of Gothenburg , Gothenburg 40530 , Sweden
| |
Collapse
|
46
|
Abstract
The mucus layer covering the gastrointestinal tract plays a critical role in maintaining a homeostatic relationship with our gut microbiota. [...].
Collapse
Affiliation(s)
- Nathalie Juge
- Quadram Institute Bioscience, The Gut Microbes and Health Institute Strategic Programme, Norwich Research Park, Norwich NR4 7UA, UK.
| |
Collapse
|