1
|
Castranova D, Kenton MI, Kraus A, Dell CW, Park JS, Venero Galanternik M, Park G, Lumbantobing DN, Dye L, Marvel M, Iben J, Taimatsu K, Pham V, Willms RJ, Blevens L, Robertson TF, Hou Y, Huttenlocher A, Foley E, Parenti LR, Frazer JK, Narayan K, Weinstein BM. The axillary lymphoid organ is an external, experimentally accessible immune organ in the zebrafish. J Exp Med 2025; 222:e20241435. [PMID: 40167600 PMCID: PMC11960710 DOI: 10.1084/jem.20241435] [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: 08/12/2024] [Revised: 01/13/2025] [Accepted: 03/03/2025] [Indexed: 04/02/2025] Open
Abstract
Lymph nodes and other secondary lymphoid organs play critical roles in immune surveillance and immune activation in mammals, but the deep internal locations of these organs make it challenging to image and study them in living animals. Here, we describe a previously uncharacterized external immune organ in the zebrafish ideally suited for studying immune cell dynamics in vivo, the axillary lymphoid organ (ALO). This small, translucent organ has an outer cortex teeming with immune cells, an inner medulla with a mesh-like network of fibroblastic reticular cells along which immune cells migrate, and a network of lymphatic vessels draining to a large adjacent lymph sac. Noninvasive high-resolution imaging of transgenically marked immune cells can be carried out in ALOs of living animals, which are readily accessible to external treatment. This newly discovered tissue provides a superb model for dynamic live imaging of immune cells and their interaction with pathogens and surrounding tissues, including blood and lymphatic vessels.
Collapse
Affiliation(s)
- Daniel Castranova
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Madeleine I. Kenton
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Aurora Kraus
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Christopher W. Dell
- Center for Molecular Microscopy, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Jong S. Park
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Marina Venero Galanternik
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Gilseung Park
- Section of Pediatric Hematology-Oncology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Daniel N. Lumbantobing
- Division of Fishes, Department of Vertebrate Zoology, National Museum of Natural History, Smithsonian Institution, Washington, DC, USA
| | - Louis Dye
- Microscopy and Imaging Core, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Miranda Marvel
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - James Iben
- Molecular Genomics Core, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Kiyohito Taimatsu
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Van Pham
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Reegan J. Willms
- Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Lucas Blevens
- Section of Pediatric Hematology-Oncology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Tanner F. Robertson
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI, USA
| | - Yiran Hou
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI, USA
| | - Anna Huttenlocher
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI, USA
| | - Edan Foley
- Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Lynne R. Parenti
- Division of Fishes, Department of Vertebrate Zoology, National Museum of Natural History, Smithsonian Institution, Washington, DC, USA
| | - J. Kimble Frazer
- Section of Pediatric Hematology-Oncology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Kedar Narayan
- Center for Molecular Microscopy, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Brant M. Weinstein
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
2
|
Wang J, Gao J, Sheng X, Tang X, Xing J, Chi H, Zhan W. Teleost Muc2 and Muc5ac: Key guardians of mucosal immunity in flounder (Paralichthys olivaceus). Int J Biol Macromol 2024; 277:134127. [PMID: 39053833 DOI: 10.1016/j.ijbiomac.2024.134127] [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: 05/07/2024] [Revised: 07/20/2024] [Accepted: 07/22/2024] [Indexed: 07/27/2024]
Abstract
Mucins secreted by mucous cells constitute a core part of the defense line against the invasion of pathogens. However, mucins' structure and immunological functions remain largely unknown in teleost fish. In this study, two typical mucins, Muc2 and Muc5ac of flounder (Paralichthys olivaceus), were cloned and their physicochemical properties, structure and conservation were analyzed. Notably, specific antibodies against flounder Muc2 and Muc5ac were developed. It was verified at the gene and protein level that Muc2 was expressed in the hindgut and gills but not in the skin, while Muc5ac was expressed in the skin and gills but not in the hindgut. After flounders were immunized by immersion with inactivated Edwardsiella tarda, Muc2 and Muc5ac were significantly upregulated at both the gene expression and protein levels, and Muc2+/Muc5ac+ mucous cells proliferated and increased secretion of Muc2 and Muc5ac. Moreover, Muc2 and Muc5ac exerted retention and clearance effects on E. tarda in a short period (within 1 dpi). These results revealed the characterization of fish mucins Muc2 and Muc5ac at the protein level and clarified the role of mucins as key guardians to maintain the mucus barrier, which advanced our understanding of teleost mucosal barrier.
Collapse
Affiliation(s)
- Jincheng Wang
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, PR China
| | - Jianliang Gao
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, PR China
| | - Xiuzhen Sheng
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, PR China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao Marine Science and Technology Center, Qingdao, Shandong 266237, PR China.
| | - Xiaoqian Tang
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, PR China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao Marine Science and Technology Center, Qingdao, Shandong 266237, PR China
| | - Jing Xing
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, PR China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao Marine Science and Technology Center, Qingdao, Shandong 266237, PR China
| | - Heng Chi
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, PR China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao Marine Science and Technology Center, Qingdao, Shandong 266237, PR China
| | - Wenbin Zhan
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, PR China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao Marine Science and Technology Center, Qingdao, Shandong 266237, PR China
| |
Collapse
|
3
|
Quintana-Hayashi MP, Thomsson Hulthe KA, Lindén SK. In vitro fish mucosal surfaces producing mucin as a model for studying host-pathogen interactions. PLoS One 2024; 19:e0308609. [PMID: 39121037 PMCID: PMC11315345 DOI: 10.1371/journal.pone.0308609] [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: 06/25/2023] [Accepted: 07/27/2024] [Indexed: 08/11/2024] Open
Abstract
Current prophylactic and disease control measures in aquaculture highlight the need of alternative strategies to prevent disease and reduce antibiotic use. Mucus covered mucosal surfaces are the first barriers pathogens encounter. Mucus, which is mainly composed of highly glycosylated mucins, has the potential to contribute to disease prevention if we can strengthen this barrier. Therefore, aim of this study was to develop and characterize fish in vitro mucosal surface models based on commercially available cell lines that are functionally relevant for studies on mucin regulation and host-pathogen interactions. The rainbow trout (Oncorhynchus mykiss) gill epithelial cell line RTgill-W1 and the embryonic cell line from Chinook salmon (Oncorhynchus tshawytscha) CHSE-214 were grown on polycarbonate membrane inserts and chemically treated to differentiate the cells into mucus producing cells. RTGill-W1 and CHSE-214 formed an adherent layer at two weeks post-confluence, which further responded to treatment with the γ-secretase inhibitor DAPT and prolonged culture by increasing the mucin production. Mucins were metabolically labelled with N-azidoacetylgalactosamine 6 h post addition to the in vitro membranes. The level of incorporated label was relatively similar between membranes based on RTgill-W1, while larger interindividual variation was observed among the CHSE in vitro membranes. Furthermore, O-glycomics of RTgill-W1 cell lysates identified three sialylated O-glycans, namely Galβ1-3(NeuAcα2-6)GalNAcol, NeuAcα-Galβ1-3GalNAcol and NeuAcα-Galβ1-3(NeuAcα2-6)GalNAcol, resembling the glycosylation present in rainbow trout gill mucin. These glycans were also present in CHSE-214. Additionally, we demonstrated binding of the fish pathogen A. salmonicida to RTgill-W1 and CHSE-214 cell lysates. Thus, these models have similarities to in vivo mucosal surfaces and can be used to investigate the effect of pathogens and modulatory components on mucin production.
Collapse
Affiliation(s)
- Macarena P. Quintana-Hayashi
- Department of Medical Biochemistry and Cell biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Kristina A. Thomsson Hulthe
- Department of Medical Biochemistry and Cell biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Sara K. Lindén
- Department of Medical Biochemistry and Cell biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
4
|
Castranova D, Kenton MI, Kraus A, Dell CW, Park JS, Galanternik MV, Park G, Lumbantobing DN, Dye L, Marvel M, Iben J, Taimatsu K, Pham V, Willms RJ, Blevens L, Robertson TF, Hou Y, Huttenlocher A, Foley E, Parenti LR, Frazer JK, Narayan K, Weinstein BM. The axillary lymphoid organ - an external, experimentally accessible immune organ in the zebrafish. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.25.605139. [PMID: 39091802 PMCID: PMC11291151 DOI: 10.1101/2024.07.25.605139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
Lymph nodes and other secondary lymphoid organs play critical roles in immune surveillance and immune activation in mammals, but the deep internal locations of these organs make it challenging to image and study them in living animals. Here, we describe a previously uncharacterized external immune organ in the zebrafish ideally suited for studying immune cell dynamics in vivo, the axillary lymphoid organ (ALO). This small, translucent organ has an outer cortex teeming with immune cells, an inner medulla with a mesh-like network of fibroblastic reticular cells along which immune cells migrate, and a network of lymphatic vessels draining to a large adjacent lymph sac. Noninvasive high-resolution imaging of transgenically marked immune cells can be carried out in the lobes of living animals, and the ALO is readily accessible to external treatment. This newly discovered tissue provides a superb model for dynamic live imaging of immune cells and their interaction with pathogens and surrounding tissues, including blood and lymphatic vessels.
Collapse
Affiliation(s)
- Daniel Castranova
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20814, USA
| | - Madeleine I. Kenton
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20814, USA
| | - Aurora Kraus
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20814, USA
| | - Christopher W. Dell
- Center for Molecular Microscopy, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA and Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Jong S. Park
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20814, USA
| | - Marina Venero Galanternik
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20814, USA
| | - Gilseung Park
- Section of Pediatric Hematology-Oncology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Daniel N. Lumbantobing
- Division of Fishes, Department of Vertebrate Zoology, National Museum of Natural History, Smithsonian Institution, Washington, DC 20560, USA
| | - Louis Dye
- Microscopy and Imaging Core, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20814, USA
| | - Miranda Marvel
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20814, USA
| | - James Iben
- Molecular Genomics Core, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20814, USA
| | - Kiyohito Taimatsu
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20814, USA
| | - Van Pham
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20814, USA
| | - Reegan J. Willms
- Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Lucas Blevens
- Section of Pediatric Hematology-Oncology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Tanner F. Robertson
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI 53706
| | - Yiran Hou
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI 53706
| | - Anna Huttenlocher
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI 53706
| | - Edan Foley
- Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Lynne R. Parenti
- Division of Fishes, Department of Vertebrate Zoology, National Museum of Natural History, Smithsonian Institution, Washington, DC 20560, USA
| | - J. Kimble Frazer
- Section of Pediatric Hematology-Oncology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Kedar Narayan
- Center for Molecular Microscopy, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA and Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Brant M. Weinstein
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20814, USA
| |
Collapse
|
5
|
Liu W, Yu H, Gurbazar D, Rinchindorj D, Kang W, Qi C, Chen H, Chang X, You H, Han Y, Li Z, R. G. A, Dong W. Anti-inflammatory effects and beneficial effects of the feed additive Urtica cannabina L. in zebrafish. PLoS One 2024; 19:e0307269. [PMID: 39018284 PMCID: PMC11253947 DOI: 10.1371/journal.pone.0307269] [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: 11/20/2023] [Accepted: 06/12/2024] [Indexed: 07/19/2024] Open
Abstract
Urtica cannabina L. (UL) has been used clinically for centuries because of its anti-inflammatory properties. This study aimed to investigate the underlying mechanisms and anti-inflammatory effects of different UL concentrations in zebrafish. To elucidate UL's anti-inflammatory properties, two inflammation zebrafish models were designed 1) by severing the zebrafish's caudal fin to assess the repairing effect of UL on the tail inflammation, and 2) by inducing lipopolysaccharides (LPS)-mediated intestinal inflammation to assess the protective and reparative effects of UL on intestinal inflammation at the histological and genetic levels. Furthermore, the effect of UL on the LPS-induced intestinal flora changes was also assessed. After caudal fin resection, a scar formed on the tail of the zebrafish, and the area of the caudal fin increased by 1.30 times as much as that of the control group (P < 0.01). Moreover, this tail scar was alleviated after 10 mg/g UL supplementation but not after 30 mg/g UL dose. LPS decreased the feed intake and body weight of the zebrafish; however, these effects were reversed after 10 and 30 mg/g doses of UL. In addition, the LPS treatment also reduced the intestinal goblet cells by 49% in the zebrafish when compared with the control, which was significantly restored after 10 and 30 mg/g UL treatments. At the genetics level, the expression of the pro-inflammatory cytokine genes (TNF-α, IL6, and IL8) showed that 10 and 30 mg/g UL doses could rescue LPS-induced expression. The gut microbiota analysis revealed changes in the abundance of four major bacterial phyla in the 10 and 30 mg/g UL-treated groups, with an increased probiotic Bacteroidota and decreased pathogenic bacteria. These results indicate that UL strongly inhibits inflammation caused by caudal fin removal and LPS-induced inflammatory changes in the zebrafish intensity, suggesting that UL is a feed additive that could be developed to improve resistance to inflammation in livestock.
Collapse
Affiliation(s)
- Wuyun Liu
- Key Laboratory of Ecological Agriculture in Horqin Sandy Land, State Ethnic Affairs Commission, Wuhan, China
- College of Agriculture, Inner Mongolia Minzu University, Tongliao, Inner Mongolia, China
- Mongolian University of Life Sciences, School of Animal science & Biotechnology, Ulaanbaatar, Mongolia
- Inner Mongolia Key Laboratory of Toxicant Monitoring and Toxicology, College of Animal Science and Technology, Inner Mongolia Minzu University, Tongliao, Inner Mongolia, China
| | - Huarong Yu
- Key Laboratory of Ecological Agriculture in Horqin Sandy Land, State Ethnic Affairs Commission, Wuhan, China
- College of Agriculture, Inner Mongolia Minzu University, Tongliao, Inner Mongolia, China
| | - D. Gurbazar
- Mongolian University of Life Sciences, School of Animal science & Biotechnology, Ulaanbaatar, Mongolia
| | - D. Rinchindorj
- Mongolian University of Life Sciences, School of Animal science & Biotechnology, Ulaanbaatar, Mongolia
| | - Wei Kang
- Inner Mongolia Key Laboratory of Toxicant Monitoring and Toxicology, College of Animal Science and Technology, Inner Mongolia Minzu University, Tongliao, Inner Mongolia, China
| | - Chelimuge Qi
- Inner Mongolia Key Laboratory of Toxicant Monitoring and Toxicology, College of Animal Science and Technology, Inner Mongolia Minzu University, Tongliao, Inner Mongolia, China
| | - Hongsong Chen
- Inner Mongolia Key Laboratory of Toxicant Monitoring and Toxicology, College of Animal Science and Technology, Inner Mongolia Minzu University, Tongliao, Inner Mongolia, China
| | - Xu Chang
- Inner Mongolia Key Laboratory of Toxicant Monitoring and Toxicology, College of Animal Science and Technology, Inner Mongolia Minzu University, Tongliao, Inner Mongolia, China
| | - Huan You
- Tongliao Animal Husbandry Development Center, Tongliao, Inner Mongolia, China
| | - Yongmei Han
- Inner Mongolia Key Laboratory of Toxicant Monitoring and Toxicology, College of Animal Science and Technology, Inner Mongolia Minzu University, Tongliao, Inner Mongolia, China
| | - Zhigang Li
- Key Laboratory of Ecological Agriculture in Horqin Sandy Land, State Ethnic Affairs Commission, Wuhan, China
- College of Agriculture, Inner Mongolia Minzu University, Tongliao, Inner Mongolia, China
| | - Ahmed R. G.
- Faculty of Science, Zoology Department, Division of Anatomy and Embryology, Beni-Suef University, Beni-Suef, Egypt
| | - Wu Dong
- Inner Mongolia Key Laboratory of Toxicant Monitoring and Toxicology, College of Animal Science and Technology, Inner Mongolia Minzu University, Tongliao, Inner Mongolia, China
| |
Collapse
|
6
|
Maritan E, Quagliariello A, Frago E, Patarnello T, Martino ME. The role of animal hosts in shaping gut microbiome variation. Philos Trans R Soc Lond B Biol Sci 2024; 379:20230071. [PMID: 38497257 PMCID: PMC10945410 DOI: 10.1098/rstb.2023.0071] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 10/10/2023] [Indexed: 03/19/2024] Open
Abstract
Millions of years of co-evolution between animals and their associated microbial communities have shaped and diversified the nature of their relationship. Studies continue to reveal new layers of complexity in host-microbe interactions, the fate of which depends on a variety of different factors, ranging from neutral processes and environmental factors to local dynamics. Research is increasingly integrating ecosystem-based approaches, metagenomics and mathematical modelling to disentangle the individual contribution of ecological factors to microbiome evolution. Within this framework, host factors are known to be among the dominant drivers of microbiome composition in different animal species. However, the extent to which they shape microbiome assembly and evolution remains unclear. In this review, we summarize our understanding of how host factors drive microbial communities and how these dynamics are conserved and vary across taxa. We conclude by outlining key avenues for research and highlight the need for implementation of and key modifications to existing theory to fully capture the dynamics of host-associated microbiomes. This article is part of the theme issue 'Sculpting the microbiome: how host factors determine and respond to microbial colonization'.
Collapse
Affiliation(s)
- Elisa Maritan
- Department of Comparative Biomedicine and Food Science, University of Padova, 35020 Padova, Italy
| | - Andrea Quagliariello
- Department of Comparative Biomedicine and Food Science, University of Padova, 35020 Padova, Italy
| | - Enric Frago
- CIRAD, UMR CBGP, INRAE, Institut Agro, IRD, Université Montpellier, 34398 Montpellier, France
| | - Tomaso Patarnello
- Department of Comparative Biomedicine and Food Science, University of Padova, 35020 Padova, Italy
| | - Maria Elena Martino
- Department of Comparative Biomedicine and Food Science, University of Padova, 35020 Padova, Italy
| |
Collapse
|
7
|
Riera-Ferrer E, Del Pozo R, Muñoz-Berruezo U, Palenzuela O, Sitjà-Bobadilla A, Estensoro I, Piazzon MC. Mucosal affairs: glycosylation and expression changes of gill goblet cells and mucins in a fish-polyopisthocotylidan interaction. Front Vet Sci 2024; 11:1347707. [PMID: 38655531 PMCID: PMC11035888 DOI: 10.3389/fvets.2024.1347707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 03/22/2024] [Indexed: 04/26/2024] Open
Abstract
Introduction Secreted mucins are highly O-glycosylated glycoproteins produced by goblet cells in mucosal epithelia. They constitute the protective viscous gel layer overlying the epithelia and are involved in pathogen recognition, adhesion and expulsion. The gill polyopisthocotylidan ectoparasite Sparicotyle chrysophrii, feeds on gilthead seabream (Sparus aurata) blood eliciting severe anemia. Methods Control unexposed and recipient (R) gill samples of gilthead seabream experimentally infected with S. chrysophrii were obtained at six consecutive times (0, 11, 20, 32, 41, and 61 days post-exposure (dpe)). In histological samples, goblet cell numbers and their intensity of lectin labelling was registered. Expression of nine mucin genes (muc2, muc2a, muc2b, muc5a/c, muc4, muc13, muc18, muc19, imuc) and three regulatory factors involved in goblet cell differentiation (hes1, elf3, agr2) was studied by qPCR. In addition, differential expression of glycosyltransferases and glycosidases was analyzed in silico from previously obtained RNAseq datasets of S. chrysophrii-infected gilthead seabream gills with two different infection intensities. Results and Discussion Increased goblet cell differentiation (up-regulated elf3 and agr2) leading to neutral goblet cell hyperplasia on gill lamellae of R fish gills was found from 32 dpe on, when adult parasite stages were first detected. At this time point, acute increased expression of both secreted (muc2a, muc2b, muc5a/c) and membrane-bound mucins (imuc, muc4, muc18) occurred in R gills. Mucins did not acidify during the course of infection, but their glycosylation pattern varied towards more complex glycoconjugates with sialylated, fucosylated and branched structures, according to lectin labelling and the shift of glycosyltransferase expression patterns. Gilthead seabream gill mucosal response against S. chrysophrii involved neutral mucus hypersecretion, which could contribute to worm expulsion and facilitate gas exchange to counterbalance parasite-induced hypoxia. Stress induced by the sparicotylosis condition seems to lead to changes in glycosylation characteristic of more structurally complex mucins.
Collapse
Affiliation(s)
| | | | | | | | | | - Itziar Estensoro
- Fish Pathology Group, Instituto de Acuicultura Torre de la Sal, Consejo Superior de Investigaciones Científicas (IATS, CSIC), Castellón, Spain
| | | |
Collapse
|
8
|
Yang JH, Park JW, Kim HS, Lee S, Yerke AM, Jaiswal YS, Williams LL, Hwang S, Moon KH. Effects of Antibiotic Residues on Fish Gut Microbiome Dysbiosis and Mucosal Barrier-Related Pathogen Susceptibility in Zebrafish Experimental Model. Antibiotics (Basel) 2024; 13:82. [PMID: 38247641 PMCID: PMC10812462 DOI: 10.3390/antibiotics13010082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 01/05/2024] [Accepted: 01/13/2024] [Indexed: 01/23/2024] Open
Abstract
The symbiotic community of microorganisms in the gut plays an important role in the health of the host. While many previous studies have been performed on the interactions between the gut microbiome and the host in mammals, studies in fish are still lacking. In this study, we investigated changes in the intestinal microbiome and pathogen susceptibility of zebrafish (Danio rerio) following chronic antibiotics exposure. The chronic antibiotics exposure assay was performed on zebrafish for 30 days using oxytetracycline (Otc), sulfamethoxazole/trimethoprim (Smx/Tmp), or erythromycin (Ery), which are antibiotics widely used in the aquaculture industry. The microbiome analysis indicated that Fusobacteria, Proteobacteria, Firmicutes, and Bacteroidetes were the dominant phyla in the gut microbiome of the zebrafish used in this study. However, in Smx/Tmp-treated zebrafish, the compositions of Fusobacteria and Proteobacteria were changed significantly, and in Ery-treated zebrafish, the compositions of Proteobacteria and Firmicutes were altered significantly. Although alpha diversity analysis showed that there was no significant difference in the richness, beta diversity analysis revealed a community imbalance in the gut microbiome of all chronically antibiotics-exposed zebrafish. Intriguingly, in zebrafish with dysbiosis in the gut microbiome, the pathogen susceptibility to Edwardsiella piscicida, a representative Gram-negative fish pathogen, was reduced. Gut microbiome imbalance resulted in a higher count of goblet cells in intestinal tissue and an upregulation of genes related to the intestinal mucosal barrier. In addition, as innate immunity was enhanced by the increased mucosal barrier, immune and stress-related gene expression in the intestinal tissue was downregulated. In this study, we provide new insight into the effect of gut microbiome dysbiosis on pathogen susceptibility.
Collapse
Affiliation(s)
- Jun Hyeok Yang
- Laboratory of Marine Microbiology, Division of Convergence of Marine Science, Korea Maritime & Ocean University, Busan 49112, Republic of Korea; (J.H.Y.); (J.W.P.); (H.S.K.)
- Department of Marine Bioscience and Environment, Korea Maritime & Ocean University, Busan 49112, Republic of Korea
| | - Jeong Woo Park
- Laboratory of Marine Microbiology, Division of Convergence of Marine Science, Korea Maritime & Ocean University, Busan 49112, Republic of Korea; (J.H.Y.); (J.W.P.); (H.S.K.)
- Department of Convergence Study on the Ocean Science and Technology, Ocean Science and Technology School, Korea Maritime & Ocean University, Busan 49112, Republic of Korea
| | - Ho Sung Kim
- Laboratory of Marine Microbiology, Division of Convergence of Marine Science, Korea Maritime & Ocean University, Busan 49112, Republic of Korea; (J.H.Y.); (J.W.P.); (H.S.K.)
- Department of Convergence Study on the Ocean Science and Technology, Ocean Science and Technology School, Korea Maritime & Ocean University, Busan 49112, Republic of Korea
| | - Seungki Lee
- National Institute of Biological Resources, Environmental Research Complex, Incheon 22689, Republic of Korea;
| | - Aaron M. Yerke
- Department of Bioinformatics and Genomics, University of North Carolina at Charlotte, Charlotte, NC 28223, USA;
| | - Yogini S. Jaiswal
- Center for Excellence in Post Harvest Technologies, North Carolina Agricultural and Technical State University, The North Carolina Research Campus, 500 Laureate Way, Kannapolis, NC 28081, USA; (Y.S.J.); (L.L.W.)
| | - Leonard L. Williams
- Center for Excellence in Post Harvest Technologies, North Carolina Agricultural and Technical State University, The North Carolina Research Campus, 500 Laureate Way, Kannapolis, NC 28081, USA; (Y.S.J.); (L.L.W.)
| | - Sungmin Hwang
- Division of Practical Research, Honam National Institute Biological Resources, Mokpo-si 58762, Republic of Korea
| | - Ki Hwan Moon
- Laboratory of Marine Microbiology, Division of Convergence of Marine Science, Korea Maritime & Ocean University, Busan 49112, Republic of Korea; (J.H.Y.); (J.W.P.); (H.S.K.)
- Department of Marine Bioscience and Environment, Korea Maritime & Ocean University, Busan 49112, Republic of Korea
- Department of Convergence Study on the Ocean Science and Technology, Ocean Science and Technology School, Korea Maritime & Ocean University, Busan 49112, Republic of Korea
| |
Collapse
|
9
|
Smith TJ, Sundarraman D, Melancon E, Desban L, Parthasarathy R, Guillemin K. A mucin-regulated adhesin determines the spatial organization and inflammatory character of a bacterial symbiont in the vertebrate gut. Cell Host Microbe 2023; 31:1371-1385.e6. [PMID: 37516109 PMCID: PMC10492631 DOI: 10.1016/j.chom.2023.07.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 05/11/2023] [Accepted: 07/06/2023] [Indexed: 07/31/2023]
Abstract
In a healthy gut, microbes are often aggregated with host mucus, yet the molecular basis for this organization and its impact on intestinal health are unclear. Mucus is a viscous physical barrier separating resident microbes from epithelia, but it also provides glycan cues that regulate microbial behaviors. Here, we describe a mucin-sensing pathway in an Aeromonas symbiont of zebrafish, Aer01. In response to the mucin-associated glycan N-acetylglucosamine, a sensor kinase regulates the expression of an aggregation-promoting adhesin we named MbpA. Upon MbpA disruption, Aer01 colonizes to normal levels but is largely planktonic and more pro-inflammatory. Increasing cell surface MbpA rescues these traits. MbpA-like adhesins are common in human-associated bacteria, and the expression of an Akkermansia muciniphila MbpA-like adhesin in MbpA-deficient Aer01 restores lumenal aggregation and reverses its pro-inflammatory character. Our work demonstrates how resident bacteria use mucin glycans to modulate behaviors congruent with host health.
Collapse
Affiliation(s)
- T Jarrod Smith
- Institute of Molecular Biology, University of Oregon, Eugene, OR, USA
| | - Deepika Sundarraman
- Department of Physics and Materials Science Institute, University of Oregon, Eugene, OR, USA
| | - Ellie Melancon
- Institute of Neuroscience, University of Oregon, Eugene, OR, USA
| | - Laura Desban
- Institute of Neuroscience, University of Oregon, Eugene, OR, USA
| | - Raghuveer Parthasarathy
- Department of Physics and Materials Science Institute, University of Oregon, Eugene, OR, USA
| | - Karen Guillemin
- Institute of Molecular Biology, University of Oregon, Eugene, OR, USA; Institute of Neuroscience, University of Oregon, Eugene, OR, USA; Humans and the Microbiome Program, Canadian Institute for Advanced Research, Toronto, ON, Canada.
| |
Collapse
|
10
|
Yue Z, Fan Z, Zhang H, Feng B, Wu C, Chen S, Ouyang J, Fan H, Weng P, Feng H, Chen S, Dong M, Xu A, Huang S. Differential roles of the fish chitinous membrane in gut barrier immunity and digestive compartments. EMBO Rep 2023; 24:e56645. [PMID: 36852962 PMCID: PMC10074124 DOI: 10.15252/embr.202256645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 02/06/2023] [Accepted: 02/09/2023] [Indexed: 03/01/2023] Open
Abstract
The chitin-based peritrophic matrix (PM) is a structure critical for both gut immunity and digestion in invertebrates. PM was traditionally considered lost in all vertebrates, but a PM-like chitinous membrane (CM) has recently been discovered in fishes, which may increase the knowledge on vertebrate gut physiology and structural evolution. Here, we show that in zebrafish, the CM affects ingestion behavior, microbial homeostasis, epithelial renewal, digestion, growth, and longevity. Young mutant fish without CM appear healthy and are able to complete their life cycle normally, but with increasing age they develop gut inflammation, resulting in gut atrophy. Unlike mammals, zebrafish have no visible gel-forming mucin layers to protect their gut epithelia, but at least in young fish, the CM is not a prerequisite for the antibacterial gut immunity. These findings provide new insights into the role of the CM in fish prosperity and its eventual loss in tetrapods. These findings may also help to improve fish health and conservation, as well as to advance the understanding of vertebrate gut physiology and human intestinal diseases.
Collapse
Affiliation(s)
- Zirui Yue
- Guangdong Key Laboratory of Pharmaceutical Functional Genes, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), State Key Laboratory of Biocontrol, School of Life SciencesSun Yat‐sen UniversityGuangdongChina#
- Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and TechnologyQingdaoChina
| | - Zhaoyu Fan
- Guangdong Key Laboratory of Pharmaceutical Functional Genes, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), State Key Laboratory of Biocontrol, School of Life SciencesSun Yat‐sen UniversityGuangdongChina#
| | - Hao Zhang
- Guangdong Key Laboratory of Pharmaceutical Functional Genes, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), State Key Laboratory of Biocontrol, School of Life SciencesSun Yat‐sen UniversityGuangdongChina#
| | - Buhan Feng
- Guangdong Key Laboratory of Pharmaceutical Functional Genes, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), State Key Laboratory of Biocontrol, School of Life SciencesSun Yat‐sen UniversityGuangdongChina#
| | - Chengyi Wu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life SciencesXiamen UniversityXiamenChina
| | - Shenghui Chen
- Guangdong Key Laboratory of Pharmaceutical Functional Genes, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), State Key Laboratory of Biocontrol, School of Life SciencesSun Yat‐sen UniversityGuangdongChina#
| | - Jihua Ouyang
- Guangdong Key Laboratory of Pharmaceutical Functional Genes, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), State Key Laboratory of Biocontrol, School of Life SciencesSun Yat‐sen UniversityGuangdongChina#
| | - Huiping Fan
- Guangdong Key Laboratory of Pharmaceutical Functional Genes, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), State Key Laboratory of Biocontrol, School of Life SciencesSun Yat‐sen UniversityGuangdongChina#
| | - Panwei Weng
- Guangdong Key Laboratory of Pharmaceutical Functional Genes, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), State Key Laboratory of Biocontrol, School of Life SciencesSun Yat‐sen UniversityGuangdongChina#
| | - Huixiong Feng
- Guangdong Key Laboratory of Pharmaceutical Functional Genes, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), State Key Laboratory of Biocontrol, School of Life SciencesSun Yat‐sen UniversityGuangdongChina#
| | - Shangwu Chen
- Guangdong Key Laboratory of Pharmaceutical Functional Genes, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), State Key Laboratory of Biocontrol, School of Life SciencesSun Yat‐sen UniversityGuangdongChina#
| | - Meiling Dong
- Guangdong Key Laboratory of Pharmaceutical Functional Genes, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), State Key Laboratory of Biocontrol, School of Life SciencesSun Yat‐sen UniversityGuangdongChina#
| | - Anlong Xu
- Guangdong Key Laboratory of Pharmaceutical Functional Genes, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), State Key Laboratory of Biocontrol, School of Life SciencesSun Yat‐sen UniversityGuangdongChina#
- School of Life SciencesBeijing University of Chinese MedicineBeijingChina
| | - Shengfeng Huang
- Guangdong Key Laboratory of Pharmaceutical Functional Genes, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), State Key Laboratory of Biocontrol, School of Life SciencesSun Yat‐sen UniversityGuangdongChina#
- Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and TechnologyQingdaoChina
| |
Collapse
|
11
|
Massaquoi MS, Kong GL, Chilin-Fuentes D, Ngo JS, Horve PF, Melancon E, Hamilton MK, Eisen JS, Guillemin K. Cell-type-specific responses to the microbiota across all tissues of the larval zebrafish. Cell Rep 2023; 42:112095. [PMID: 36787219 PMCID: PMC10423310 DOI: 10.1016/j.celrep.2023.112095] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 08/22/2022] [Accepted: 01/25/2023] [Indexed: 02/15/2023] Open
Abstract
Animal development proceeds in the presence of intimate microbial associations, but the extent to which different host cells across the body respond to resident microbes remains to be fully explored. Using the vertebrate model organism, the larval zebrafish, we assessed transcriptional responses to the microbiota across the entire body at single-cell resolution. We find that cell types across the body, not limited to tissues at host-microbe interfaces, respond to the microbiota. Responses are cell-type-specific, but across many tissues the microbiota enhances cell proliferation, increases metabolism, and stimulates a diversity of cellular activities, revealing roles for the microbiota in promoting developmental plasticity. This work provides a resource for exploring transcriptional responses to the microbiota across all cell types of the vertebrate body and generating new hypotheses about the interactions between vertebrate hosts and their microbiota.
Collapse
Affiliation(s)
- Michelle S Massaquoi
- Institute of Molecular Biology, University of Oregon, 1318 Franklin Boulevard, Eugene, OR 97403, USA; Thermo Fisher Scientific, 29851 Willow Creek Road, Eugene, OR 97402, USA; Thermo Fisher Scientific, 22025 20th Avenue SE, Bothell, WA 98021, USA
| | - Garth L Kong
- Institute of Molecular Biology, University of Oregon, 1318 Franklin Boulevard, Eugene, OR 97403, USA
| | - Daisy Chilin-Fuentes
- Institute of Molecular Biology, University of Oregon, 1318 Franklin Boulevard, Eugene, OR 97403, USA
| | - Julia S Ngo
- Institute of Molecular Biology, University of Oregon, 1318 Franklin Boulevard, Eugene, OR 97403, USA
| | - Patrick F Horve
- Institute of Molecular Biology, University of Oregon, 1318 Franklin Boulevard, Eugene, OR 97403, USA
| | - Ellie Melancon
- Institute of Neuroscience, University of Oregon, 1254 University of Oregon, Eugene, OR 97403, USA
| | - M Kristina Hamilton
- Institute of Molecular Biology, University of Oregon, 1318 Franklin Boulevard, Eugene, OR 97403, USA; Institute of Neuroscience, University of Oregon, 1254 University of Oregon, Eugene, OR 97403, USA; Thermo Fisher Scientific, 29851 Willow Creek Road, Eugene, OR 97402, USA
| | - Judith S Eisen
- Institute of Molecular Biology, University of Oregon, 1318 Franklin Boulevard, Eugene, OR 97403, USA; Institute of Neuroscience, University of Oregon, 1254 University of Oregon, Eugene, OR 97403, USA
| | - Karen Guillemin
- Institute of Molecular Biology, University of Oregon, 1318 Franklin Boulevard, Eugene, OR 97403, USA; Humans and the Microbiome Program, CIFAR, Toronto, ON M5G 1M1, Canada.
| |
Collapse
|
12
|
Adade EE, Stevick RJ, Pérez-Pascual D, Ghigo JM, Valm AM. Gnotobiotic zebrafish microbiota display inter-individual variability affecting host physiology. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.01.526612. [PMID: 36778358 PMCID: PMC9915576 DOI: 10.1101/2023.02.01.526612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Gnotobiotic animal models reconventionalized under controlled laboratory conditions with multi-species bacterial communities are commonly used to study host-microbiota interactions under presumably more reproducible conditions than conventional animals. The usefulness of these models is however limited by inter-animal variability in bacterial colonization and our general lack of understanding of the inter-individual fluctuation and spatio-temporal dynamics of microbiota assemblies at the micron to millimeter scale. Here, we show underreported variability in gnotobiotic models by analyzing differences in gut colonization efficiency, bacterial composition, and host intestinal mucus production between conventional and gnotobiotic zebrafish larvae re-conventionalized with a mix of 9 bacteria isolated from conventional microbiota. Despite similar bacterial community composition, we observed high variability in the spatial distribution of bacteria along the intestinal tract in the reconventionalized model. We also observed that, whereas bacteria abundance and intestinal mucus per fish were not correlated, reconventionalized fish had lower intestinal mucus compared to conventional animals, indicating that the stimulation of mucus production depends on the microbiota composition. Our findings, therefore, suggest that variable colonization phenotypes affect host physiology and impact the reproducibility of experimental outcomes in studies that use gnotobiotic animals. This work provides insights into the heterogeneity of gnotobiotic models and the need to accurately assess re-conventionalization for reproducibility in host-microbiota studies.
Collapse
Affiliation(s)
- Emmanuel E. Adade
- Department of Biological Sciences, State University of New York at Albany, Albany, NY 12222, USA
- The RNA Institute, State University of New York at Albany, Albany, NY 12222, USA
| | - Rebecca J. Stevick
- Institut Pasteur, Université de Paris Cité, CNRS UMR 6047, Genetics of Biofilms Laboratory, Paris F-75015, France
| | - David Pérez-Pascual
- Institut Pasteur, Université de Paris Cité, CNRS UMR 6047, Genetics of Biofilms Laboratory, Paris F-75015, France
| | - Jean-Marc Ghigo
- Institut Pasteur, Université de Paris Cité, CNRS UMR 6047, Genetics of Biofilms Laboratory, Paris F-75015, France
| | - Alex M. Valm
- Department of Biological Sciences, State University of New York at Albany, Albany, NY 12222, USA
- The RNA Institute, State University of New York at Albany, Albany, NY 12222, USA
| |
Collapse
|
13
|
Sourani Z, Shirian S, Shafiei S, Mosayebi N, Nematollahi A. Modulation of Immune-Related Gene Expressions in Zebrafish (Danio rerio) by Dietary Purslane (Portulaca oleracea) Extract. MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2023; 25:214-221. [PMID: 36609893 DOI: 10.1007/s10126-022-10195-z] [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: 07/08/2022] [Accepted: 12/29/2022] [Indexed: 06/17/2023]
Abstract
To promote fish's immunity against pathogens in the aquaculture industry, fish dietary fortification with additives or compounds has increasingly attracted attention. In the present study, zebrafish (Danio rerio) was used as an animal model to investigate the effects of purslane, Portulaca oleracea, extract (PE) on the relative expression level of some immune-related genes. A total of 300 zebrafish were randomly divided into four treatment groups and fed for 8 weeks with the basal diets supplemented with 0.5, 1, 1.5, and 2% of PE. The control group was fed with a basal diet without PE. At the end of 8 weeks, the mRNA expression levels of interleukin 1-beta (IL-1β), interleukin 10 (IL-10), transforming growth factor-beta (TGF-β), tumor necrosis factor-alpha (TNF-α), superoxide dismutase (SOD), and lysozyme (LYZ) in the fish were evaluated. The results showed that the mRNA expression level of IL-1β was significantly upregulated in the fish fed with 1 and 2% PE compared to the control group (p < 0.05). Moreover, the evaluation of the mRNA expression level of TGF-β was significantly increased in a dose-dependent manner in the 1.5 and 2% fed groups compared to the control group (p < 0.05). However, the IL-10 was significantly downregulated in all treated groups compared to the control group (p < 0.05). The expression of the TNF-α gene was not affected amongst all groups by the inclusion of PE in the zebrafish diet (p > 0.05). Based on the results, the diet supplemented with 1.5 and 2% PE significantly upregulated the mRNA expression levels of LYZ and SOD, respectively, compared to the control group (p < 0.05). In conclusion, dietary inclusion of PE may result in beneficial effects on some immune responses via upregulation of some immune genes in zebrafish.
Collapse
Affiliation(s)
- Zahra Sourani
- Department of Pathology, Faculty of Veterinary Medicine, Shahrekord University, Shahrekord, Iran
| | - Sadegh Shirian
- Department of Pathology, Faculty of Veterinary Medicine, Shahrekord University, Shahrekord, Iran
| | - Shafigh Shafiei
- Department of Food Hygiene and Quality Control, Faculty of Veterinary Medicine, Shahrekord University, Shahrekord, Iran.
| | - Nadia Mosayebi
- Department of Food Hygiene and Quality Control, Faculty of Veterinary Medicine, Shahrekord University, Shahrekord, Iran
| | - Amin Nematollahi
- Department of Food Hygiene and Quality Control, Faculty of Veterinary Medicine, Shahrekord University, Shahrekord, Iran
| |
Collapse
|
14
|
Leal E, Angotzi AR, Gregório SF, Ortiz-Delgado JB, Rotllant J, Fuentes J, Tafalla C, Cerdá-Reverter JM. Role of the melanocortin system in zebrafish skin physiology. FISH & SHELLFISH IMMUNOLOGY 2022; 130:591-601. [PMID: 36150411 DOI: 10.1016/j.fsi.2022.09.036] [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: 07/11/2022] [Revised: 09/09/2022] [Accepted: 09/15/2022] [Indexed: 06/16/2023]
Abstract
The agouti-signaling protein (ASIP) acts as both a competitive antagonist and inverse agonist of melanocortin receptors which regulate dorsal-ventral pigmentation patterns in fish. However, the potential role of ASIP in the regulation of additional physiological pathways in the skin is unknown. The skin plays a crucial role in the immune function, acting as a physical limitation against infestation and also as a chemical barrier due to its ability to synthesize and secrete mucus and many immune effector proteins. In this study, the putative role of ASIP in regulating the immune system of skin has been explored using a transgenic zebrafish model overexpressing the asip1 gene (ASIPzf). Initially, the structural changes in skin induced by asip1 overexpression were studied, revealing that the ventral skin of ASIPzf was thinner than that of wild type (WT) animals. A moderate hypertrophy of mucous cells was also found in ASIPzf. Histochemical studies showed that transgenic animals appear to compensate for the lower number of cell layers by modifying the mucus composition and increasing lectin affinity and mucin content in order to maintain or improve protection against microorganism adhesion. ASIPzf also exhibit higher protein concentration under crowding conditions suggesting an increased mucus production under stressful conditions. Exposure to bacterial lipopolysaccharide (LPS) showed that ASIPzf exhibit a faster pro-inflammatory response and increased mucin expression yet severe skin injures and a slight increase in mortality was observed. Electrophysiological measurements show that the ASIP1 genotype exhibits reduced epithelial resistance, an indicator of reduced tissue integrity and barrier function. Overall, not only are ASIP1 animals more prone to infiltration and subsequent infections due to reduced skin epithelial integrity, but also display an increased inflammatory response that can lead to increased skin sensitivity to external infections.
Collapse
Affiliation(s)
- E Leal
- Department of Fish Physiology and Biotechnology, Institute of Aquiculture de Torre de la Sal, IATS-CSIC, 12595, Castellon, Spain.
| | - A R Angotzi
- Department of Fish Physiology and Biotechnology, Institute of Aquiculture de Torre de la Sal, IATS-CSIC, 12595, Castellon, Spain
| | - S F Gregório
- Centre of Marine Sciences (CCMar), Universidade do Algarve Campus de Gambelas, 8005-139, Faro, Portugal
| | - J B Ortiz-Delgado
- Instituto de Ciencias Marinas de Andalucía-ICMAN, CSIC Campus Universitario Río San Pedro, 11510, Puerto Real, Cádiz, Spain
| | - J Rotllant
- Instituto de Investigaciones Marinas (IIM), CSIC, 36208, Vigo, Spain
| | - J Fuentes
- Centre of Marine Sciences (CCMar), Universidade do Algarve Campus de Gambelas, 8005-139, Faro, Portugal
| | - C Tafalla
- Animal Health Research Center (CISA-INIA-CSIC), Valdeolmos, 28130, Madrid, Spain
| | - J M Cerdá-Reverter
- Department of Fish Physiology and Biotechnology, Institute of Aquiculture de Torre de la Sal, IATS-CSIC, 12595, Castellon, Spain.
| |
Collapse
|
15
|
Huang Y, Li L, Rong YS. JiangShi(僵尸): a widely distributed Mucin-like protein essential for Drosophila development. G3 GENES|GENOMES|GENETICS 2022; 12:6589892. [PMID: 35595239 PMCID: PMC9339309 DOI: 10.1093/g3journal/jkac126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 05/11/2022] [Indexed: 11/30/2022]
Abstract
Epithelia exposed to elements of the environment are protected by a mucus barrier in mammals. This barrier also serves to lubricate during organ movements and to mediate substance exchanges between the environmental milieu and internal organs. A major component of the mucus barrier is a class of glycosylated proteins called Mucin. Mucin and mucin-related proteins are widely present in the animal kingdom. Mucin mis-regulation has been reported in many diseases such as cancers and ones involving the digestive and respiratory tracts. Although the biophysical properties of isolated Mucins have been extensively studied, in vivo models remain scarce for the study of their functions and regulations. Here, we characterize the Mucin-like JiangShi protein and its mutations in the fruit fly Drosophila. JiangShi is an extracellular glycoprotein with domain features reminiscent of mammalian nonmembranous Mucins, and one of the most widely distributed Mucin-like proteins studied in Drosophila. Both loss and over-production of JiangShi lead to terminal defects in adult structures and organismal death. Although the physiological function of JiangShi remains poorly defined, we present a genetically tractable model system for the in vivo studies of Mucin-like molecules.
Collapse
Affiliation(s)
- Yueping Huang
- School of Life Sciences, Sun Yat-sen University , Guangzhou 510275, China
- Hengyang College of Medicine, University of South China , Hengyang 421009, China
| | - LingLing Li
- School of Life Sciences, Sun Yat-sen University , Guangzhou 510275, China
| | - Yikang S Rong
- Hengyang College of Medicine, University of South China , Hengyang 421009, China
| |
Collapse
|
16
|
Sharba S, Sundh H, Sundell K, Benktander J, Santos L, Birchenough G, Lindén SK. Rainbow trout gastrointestinal mucus, mucin production, mucin glycosylation and response to lipopolysaccharide. FISH & SHELLFISH IMMUNOLOGY 2022; 122:181-190. [PMID: 35077869 DOI: 10.1016/j.fsi.2022.01.031] [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: 10/29/2021] [Revised: 01/19/2022] [Accepted: 01/19/2022] [Indexed: 06/14/2023]
Abstract
Mucus, whereof the highly glycosylated mucins are a major component, protects the epithelial mucosal surfaces. The aim of this study was to characterize the rainbow trout (Oncorhynchus mykiss) gastrointestinal mucus barrier function, mucin production, glycosylation and response to lipopolysaccharide. Both gastric and intestinal mucus was thick and impenetrable to bacteria-sized beads ex vivo. The secreted mucus covering the gastric epithelium predominantly contained sialylated mucins. Plume-like structures emerging from the gastric pits were both sialylated and fucosylated, indicating heterogeneity in gastric mucus secreted by the surface mucus cells and gland secretory cells, whereas intestinal mucus appeared more homogenous. In vivo metabolic mucin labelling revealed regional differences in mucin production and basal to apical transport, while lipopolysaccharide stimulation increased the rate of mucin production and basal to apical transport in both stomach and intestine. Using mass spectrometry, 34 mucin O-glycans were identified, with ∼70% of the relative abundance being sialylated, ∼40% di-sialylated and 20-25% fucosylated. No effects of lipopolysaccharide treatment were apparent regarding O-glycan repertoires, relative abundance of components, size distribution or core structures. Thus, the mucus production and organization differ between epithelial sites but provide a barrier to bacteria in both stomach and intestine. Furthermore, mucin production and basal to apical transport was stimulated by lipopolysaccharide in all regions, suggesting a mechanism to combat infections.
Collapse
Affiliation(s)
- Sinan Sharba
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Box 440, Medicinaregatan 9A, 405 30, Gothenburg, Sweden
| | - Henrik Sundh
- Department of Biological and Environmental Sciences, University of Gothenburg, 405 30, Gothenburg, Sweden
| | - Kristina Sundell
- Department of Biological and Environmental Sciences, University of Gothenburg, 405 30, Gothenburg, Sweden
| | - John Benktander
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Box 440, Medicinaregatan 9A, 405 30, Gothenburg, Sweden
| | - Licinia Santos
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Box 440, Medicinaregatan 9A, 405 30, Gothenburg, Sweden
| | - George Birchenough
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Box 440, Medicinaregatan 9A, 405 30, Gothenburg, Sweden
| | - Sara K Lindén
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Box 440, Medicinaregatan 9A, 405 30, Gothenburg, Sweden.
| |
Collapse
|
17
|
A cell atlas of microbe-responsive processes in the zebrafish intestine. Cell Rep 2022; 38:110311. [PMID: 35108531 DOI: 10.1016/j.celrep.2022.110311] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 10/28/2021] [Accepted: 01/07/2022] [Indexed: 02/08/2023] Open
Abstract
Gut microbial products direct growth, differentiation, and development in animal hosts. However, we lack system-wide understanding of cell-specific responses to the microbiome. We profiled cell transcriptomes from the intestine, and associated tissue, of zebrafish larvae raised in the presence or absence of a microbiome. We uncovered extensive cellular heterogeneity in the conventional zebrafish intestinal epithelium, including previously undescribed cell types with known mammalian homologs. By comparing conventional to germ-free profiles, we mapped microbial impacts on transcriptional activity in each cell population. We revealed intricate degrees of cellular specificity in host responses to the microbiome that included regulatory effects on patterning and on metabolic and immune activity. For example, we showed that the absence of microbes hindered pro-angiogenic signals in the developing vasculature, causing impaired intestinal vascularization. Our work provides a high-resolution atlas of intestinal cellular composition in the developing fish gut and details the effects of the microbiome on each cell type.
Collapse
|
18
|
Fujii S, Kobayashi S, Tettey PA, Lisdiana L, Saito H, Kawai K, Sambongi Y. Transcriptome analysis of Chironomus sulfurosus larvae living in acidic environments: Insights into molecular mechanisms for acid tolerance. JOURNAL OF INSECT PHYSIOLOGY 2021; 133:104288. [PMID: 34343520 DOI: 10.1016/j.jinsphys.2021.104288] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 07/26/2021] [Accepted: 07/27/2021] [Indexed: 06/13/2023]
Abstract
Larvae of chironomid Chironomus sulfurosus mainly live in acidic rivers near hot springs, suggesting that they naturally select acidic environments as preferred habitats. Here we showed that C. sulfurosus larvae moved toward acidic areas and stayed alive on agar gels with a pH gradient of H2SO4, and the body fluid pH of the homogenized larvae was near neutral even acclimated under the acidic conditions, indicating mechanisms for acid tolerance. In order to gain insights into this mechanism at the molecular level, de novo RNA-seq analysis was performed on C. sulfurosus larvae. As a result, 1,208 genes were found to be significantly up-regulated in larvae acclimated at pH 2.0 compared to controls at pH 7.0. Among the up-regulated genes, ones encoding cuticle proteins, peritrophic matrix proteins, mucus-forming proteins, F-type ATPase subunits, glutathione S transferases, β-1,3-D-glucan synthetase, hemoglobin, and cytochrome P450 were identified. This transcriptome analysis in conjunction with behavioral and biochemical assays expands our knowledge of gene expression in C. sulfurosus larvae living in acidic environments, which will provide a basis for further studies to elucidate the molecular mechanisms for acid tolerance employed by organisms in nature.
Collapse
Affiliation(s)
- Sotaro Fujii
- Graduate School of Integrated Sciences for Life, Hiroshima University, Kagamiyama, Higashi-Hiroshima, Hiroshima 739-8528, Japan.
| | - Satoru Kobayashi
- Graduate School of Biosphere Science, Hiroshima University, Kagamiyama, Higashi-Hiroshima, Hiroshima 739-8528, Japan
| | - Pamela A Tettey
- Graduate School of Integrated Sciences for Life, Hiroshima University, Kagamiyama, Higashi-Hiroshima, Hiroshima 739-8528, Japan
| | - Lisa Lisdiana
- Department of Biology, Universitas Negeri Surabaya, Kampus Unesa Ketintang, Surabaya 60231, Indonesia
| | - Hidetoshi Saito
- Graduate School of Integrated Sciences for Life, Hiroshima University, Kagamiyama, Higashi-Hiroshima, Hiroshima 739-8528, Japan
| | - Koichiro Kawai
- Graduate School of Integrated Sciences for Life, Hiroshima University, Kagamiyama, Higashi-Hiroshima, Hiroshima 739-8528, Japan
| | - Yoshihiro Sambongi
- Graduate School of Integrated Sciences for Life, Hiroshima University, Kagamiyama, Higashi-Hiroshima, Hiroshima 739-8528, Japan.
| |
Collapse
|
19
|
Wen J, Mercado GP, Volland A, Doden HL, Lickwar CR, Crooks T, Kakiyama G, Kelly C, Cocchiaro JL, Ridlon JM, Rawls JF. Fxr signaling and microbial metabolism of bile salts in the zebrafish intestine. SCIENCE ADVANCES 2021; 7:eabg1371. [PMID: 34301599 PMCID: PMC8302129 DOI: 10.1126/sciadv.abg1371] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Accepted: 06/07/2021] [Indexed: 05/02/2023]
Abstract
Bile salt synthesis, secretion into the intestinal lumen, and resorption in the ileum occur in all vertebrate classes. In mammals, bile salt composition is determined by host and microbial enzymes, affecting signaling through the bile salt-binding transcription factor farnesoid X receptor (Fxr). However, these processes in other vertebrate classes remain poorly understood. We show that key components of hepatic bile salt synthesis and ileal transport pathways are conserved and under control of Fxr in zebrafish. Zebrafish bile salts consist primarily of a C27 bile alcohol and a C24 bile acid that undergo multiple microbial modifications including bile acid deconjugation that augments Fxr activity. Using single-cell RNA sequencing, we provide a cellular atlas of the zebrafish intestinal epithelium and uncover roles for Fxr in transcriptional and differentiation programs in ileal and other cell types. These results establish zebrafish as a nonmammalian vertebrate model for studying bile salt metabolism and Fxr signaling.
Collapse
Affiliation(s)
- Jia Wen
- Department of Molecular Genetics and Microbiology, Duke Microbiome Center, Duke University School of Medicine, Durham, NC, USA
| | - Gilberto Padilla Mercado
- Department of Molecular Genetics and Microbiology, Duke Microbiome Center, Duke University School of Medicine, Durham, NC, USA
| | - Alyssa Volland
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana Champaign, Urbana, IL, USA
| | - Heidi L Doden
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana Champaign, Urbana, IL, USA
- Department of Animal Sciences, University of Illinois at Urbana Champaign, Urbana, IL, USA
| | - Colin R Lickwar
- Department of Molecular Genetics and Microbiology, Duke Microbiome Center, Duke University School of Medicine, Durham, NC, USA
| | - Taylor Crooks
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana Champaign, Urbana, IL, USA
| | - Genta Kakiyama
- Department of Internal Medicine, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Cecelia Kelly
- Department of Molecular Genetics and Microbiology, Duke Microbiome Center, Duke University School of Medicine, Durham, NC, USA
| | - Jordan L Cocchiaro
- Department of Molecular Genetics and Microbiology, Duke Microbiome Center, Duke University School of Medicine, Durham, NC, USA
| | - Jason M Ridlon
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana Champaign, Urbana, IL, USA.
- Department of Animal Sciences, University of Illinois at Urbana Champaign, Urbana, IL, USA
- Division of Nutritional Sciences, University of Illinois at Urbana Champaign, Urbana, IL, USA
- Cancer Center of Illinois, Urbana, IL, USA
| | - John F Rawls
- Department of Molecular Genetics and Microbiology, Duke Microbiome Center, Duke University School of Medicine, Durham, NC, USA.
| |
Collapse
|
20
|
Owen JP, Kelsh RN. A suitable anaesthetic protocol for metamorphic zebrafish. PLoS One 2021; 16:e0246504. [PMID: 33667238 PMCID: PMC7935316 DOI: 10.1371/journal.pone.0246504] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 01/21/2021] [Indexed: 02/01/2023] Open
Abstract
Zebrafish are frequently used as a means to investigate development. These studies increasingly require repeated anaesthesia of zebrafish during juvenile (i.e. metamorphic) stages. The effects of anaesthesia during this time remain poorly studied. The aim of this study was to develop a reliable method that can be used for frequently repeated anaesthesia during juvenile stages. Initially, we assessed different concentrations of MS-222, the most commonly used fish anaesthetic, for 30 minute anaesthesia with recovery. We showed that suitable MS-222 doses could be identified for the smallest (7mm) and largest (20mm) fish. However, we found that juvenile fish within a specific metamorphic window (sized between 8-16 mm) were vulnerable to MS-222 and no standard concentration of MS-222 provided reliable anaesthesia under these conditions. Hence we focussed our efforts on identifying a protocol for these stages. We tested six different published anaesthesia protocols P1-P6 where P1, P2 corresponds to 0.01% MS-222, P3, P4: 0.085% 2-phenoxyethanol and P5, P6: 0.00025%/0.0050% Propofol/Lidocaine. In protocols P1, P3, P5 fish were maintained by immersion, whilst in P2, P4 and P6: fish were maintained on an anaesthetic-doused cotton-pad. We assessed reliable anaesthesia using 10 fish for 10 minutes, with full recovery. Our data allowed us to eliminate two of these protocols as unsuitable for short term anaesthesia with recovery of juvenile fish. Extending these studies to explore repeated anaesthesia at 4 day intervals for 20 days under the remaining four protocols, we showed that P1 and P4 were both suitable for repeated anaesthesia, and that P4 was most suitable for imaging. We confirmed that P4 remained suitable when the frequency of anaesthesia was increased to every 2 days. We conclude that this protocol provides a refinement to the current protocol for repeated anaesthesia with recovery of juvenile zebrafish in the vulnerable metamorphic window.
Collapse
Affiliation(s)
- Jennifer P. Owen
- Department of Biology and Biochemistry, University of Bath, Bath, United Kingdom
| | - Robert N. Kelsh
- Department of Biology and Biochemistry, University of Bath, Bath, United Kingdom
| |
Collapse
|
21
|
Coleman OI, Haller D. Microbe-Mucus Interface in the Pathogenesis of Colorectal Cancer. Cancers (Basel) 2021; 13:cancers13040616. [PMID: 33557139 PMCID: PMC7913824 DOI: 10.3390/cancers13040616] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/18/2021] [Accepted: 01/27/2021] [Indexed: 12/24/2022] Open
Abstract
Overlying gastrointestinal epithelial cells is the transparent mucus layer that separates the lumen from the host. The dynamic mucus layer serves to lubricate the mucosal surface, to protect underlying epithelial cells, and as a transport medium between luminal contents and epithelial cells. Furthermore, it provides a habitat for commensal bacteria and signals to the underlying immune system. Mucins are highly glycosylated proteins, and their glycocode is tissue-specific and closely linked to the resident microbiota. Aberrant mucin expression and glycosylation are linked to chronic inflammation and gastrointestinal cancers, including colorectal cancer (CRC). Aberrant mucus production compromises the mucus layer and allows bacteria to come into close contact with the intestinal epithelium, potentially triggering unfavorable host responses and the subsequent development of tumors. Here, we review our current understanding of the interaction between the intestinal microbiota and mucus in healthy and CRC subjects. Deep knowledge of the intricate mechanisms of microbe-mucus interactions may contribute to the development of novel treatment strategies for CRC, in which a dysfunctional mucus layer is observed.
Collapse
Affiliation(s)
- Olivia I. Coleman
- Department of Nutrition and Immunology, School of Life Sciences, Technical University of Munich, 85354 Freising, Germany;
- Correspondence: ; Tel.: +49-08161-71-2375
| | - Dirk Haller
- Department of Nutrition and Immunology, School of Life Sciences, Technical University of Munich, 85354 Freising, Germany;
- ZIEL—Institute for Food & Health, Technical University of Munich, 85354 Freising, Germany
| |
Collapse
|
22
|
Chen D, Peng S, Chen D, Yang F, Liu J, Wang J, Liu Q, Huang X, Ouyang P, Wang K, Li Z, Geng Y. Low lethal doses of Streptococcus iniae caused enteritis in Siberian sturgeon (Acipenser baerii). FISH & SHELLFISH IMMUNOLOGY 2020; 104:654-662. [PMID: 32561456 DOI: 10.1016/j.fsi.2020.06.020] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 06/05/2020] [Accepted: 06/08/2020] [Indexed: 06/11/2023]
Abstract
In aquaculture, the incidence of enteritis due to Streptococcus iniae infection in Siberian sturgeon (Acipenser baerii) has increased in recent years. The pathogenesis of S. iniae is largely unknown due to the paucity of experimental studies on fish intestinal inflammation. In this study, S. iniae infection of A. baerii juveniles was induced by anal intubation of 0.15 mL at a low lethal dose (2 × 107 CFU/mL). Intestinal pathology and gene expression studies were conducted within 10 days of the experiment. Histopathological examination showed severe intestinal lesions, inflammatory cell infiltration, intestinal submucosa edema, epithelial cell shedding and necrosis. Predominant symptoms of exudative inflammation, metamorphic inflammation and proliferative inflammation on days 1-3, 4-6, and 7-10 post infection were shown, respectively. Ultrastructural observations also revealed fractured microvilli and shedding on days 4-6. Intestinal villi gradually repaired during the subsequent 7-10 days post infection. Expression of the pro-inflammatory cytokines, tumor necrosis factor and interleukin 1β were up-regulated on days 1-3 followed by a significant decrease on day 5, ultimately reaching control levels on day 10 post infection. A similar pattern was shown in mucus cells, involving mucin secretion and expression of the mucin encoding gene, Mucin-2. These results showed the cellular response to S. iniae infection associated with inflammatory genes expression in the Siberian sturgeon.
Collapse
Affiliation(s)
- Defang Chen
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, PR China
| | - Shuang Peng
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, PR China
| | - Daiyu Chen
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, PR China
| | - Fei Yang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, PR China
| | - Jiaxi Liu
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, PR China
| | - Jun Wang
- Key Laboratory of Sichuan Province for Fishes Conservation and Utilization in the Upper Reaches of the Yangtze River, Neijiang Normal University, Neijiang, Sichuan, PR China
| | - Qiwei Liu
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, PR China
| | - Xiaoli Huang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, PR China
| | - Ping Ouyang
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, PR China
| | - Kaiyu Wang
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, PR China
| | - Zhiqiong Li
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, PR China
| | - Yi Geng
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, PR China.
| |
Collapse
|
23
|
Gong X, Jiang S, Tian H, Xiang D, Zhang J. Polyphenols in the Fermentation Liquid of Dendrobium candidum Relieve Intestinal Inflammation in Zebrafish Through the Intestinal Microbiome-Mediated Immune Response. Front Immunol 2020; 11:1542. [PMID: 32765533 PMCID: PMC7379839 DOI: 10.3389/fimmu.2020.01542] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 06/11/2020] [Indexed: 01/04/2023] Open
Abstract
Previous studies of Dendrobium candidum (D. candidum), which is mainly distributed in tropical areas, have mainly focused on its functional polysaccharide; the effects of D. candidum polyphenols, the chemical composition of which may be improved by fermentation, have received limited attention, especially in in vivo models, which inevitably involve interactions with intestinal microorganisms. To address this challenge, metagenomic and metabolomic techniques, were applied, and immune factors and mucosal barrier-related proteins were determined to reveal the effects of fermented D. candidum polyphenols (FDC) on intestinal inflammation induced by oxazolone in zebrafish. The results showed that fermentation significantly changed the chemical composition of D. candidum and that FDC significantly improved the intestinal immune index. After the 21st day of FDC intervention, the abundance of Lactobacillus, Faecalibacterium, and Rummeliibacillus increased, but the abundance of the genera Shewanella, Geodermatophilus, Peptostreptococcaceae, and Mycobacterium decreased. At the same time, FDC significantly increased intestinal short-chain fatty acids (SCFAs). In addition, network analysis based on multi-omics indicated that FDC intake leads to changes in intestinal microbiota and intestinal metabolites, resulting in enhanced host immune function. These results indicate that FDC can improve intestinal health by regulating the intestinal microbiota and its metabolites to treat intestinal inflammation and regulate the host immune system. The present research improved our understanding of the utilization of D. candidum polyphenols and provided new evidence for the impacts of fermented D. candidum on host health.
Collapse
Affiliation(s)
- Xiaoyue Gong
- College of Food Science and Engineering, Hainan University, Haikou, China
| | - Shuaiming Jiang
- College of Food Science and Engineering, Hainan University, Haikou, China
| | - Haiyan Tian
- College of Food Science and Engineering, Hainan University, Haikou, China
| | - Dong Xiang
- College of Food Science and Engineering, Hainan University, Haikou, China.,Key Laboratory of Food Nutrition and Functional Food in Hainan Province, Hainan University, Haikou, China
| | - Jiachao Zhang
- College of Food Science and Engineering, Hainan University, Haikou, China
| |
Collapse
|
24
|
Qiang L, Arabeyyat ZH, Xin Q, Paunov VN, Dale IJF, Lloyd Mills RI, Rotchell JM, Cheng J. Silver Nanoparticles in Zebrafish ( Danio rerio) Embryos: Uptake, Growth and Molecular Responses. Int J Mol Sci 2020; 21:ijms21051876. [PMID: 32182933 PMCID: PMC7084859 DOI: 10.3390/ijms21051876] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Revised: 02/27/2020] [Accepted: 03/03/2020] [Indexed: 12/27/2022] Open
Abstract
Silver nanoparticles (AgNPs) are widely used in commercial applications as antimicrobial agents, but there have recently been increasing concerns raised about their possible environmental and health impacts. In this study, zebrafish embryos were exposed to two sizes of AgNP, 4 and 10 nm, through a continuous exposure from 4 to 96 h post-fertilisation (hpf), to study their uptake, impact and molecular defense responses. Results showed that zebrafish embryos were significantly impacted by 72 hpf when continuously exposed to 4 nm AgNPs. At concentrations above 0.963 mg/L, significant in vivo uptake and delayed yolk sac absorption was evident; at 1.925 mg/L, significantly reduced body length was recorded compared to control embryos. Additionally, 4 nm AgNP treatment at the same concentration resulted in significantly upregulated hypoxia inducible factor 4 (HIF4) and peroxisomal membrane protein 2 (Pxmp2) mRNA expression in exposed embryos 96 hpf. In contrast, no significant differences in terms of larvae body length, yolk sac absorption or gene expression levels were observed following exposure to 10 nm AgNPs. These results demonstrated that S4 AgNPs are available for uptake, inducing developmental (measured as body length and yolk sac area) and transcriptional (specifically HIF4 and Pxmp2) perturbations in developing embryos. This study suggests the importance of particle size as one possible factor in determining the developmental toxicity of AgNPs in fish embryos.
Collapse
Affiliation(s)
- Liyuan Qiang
- State Key Laboratory of Estuarine and Coastal Research, East China Normal University, Shanghai 200062, China; (L.Q.); (Q.X.)
| | - Zeinab H. Arabeyyat
- Department of Marine Biology, the University of Jordan, Aqaba branch, Aqaba 77111, Jordan;
| | - Qi Xin
- State Key Laboratory of Estuarine and Coastal Research, East China Normal University, Shanghai 200062, China; (L.Q.); (Q.X.)
| | - Vesselin N. Paunov
- Department of Chemistry and Biochemistry, University of Hull, Cottingham Road, Hull HU6 7RX, UK;
| | - Imogen J. F. Dale
- School of Biological, Biomedical, and Environmental Sciences, University of Hull, Cottingham Road, Hull HU6 7RX, UK; (I.J.F.D.); (R.I.L.M.)
| | - Richard I. Lloyd Mills
- School of Biological, Biomedical, and Environmental Sciences, University of Hull, Cottingham Road, Hull HU6 7RX, UK; (I.J.F.D.); (R.I.L.M.)
| | - Jeanette M. Rotchell
- School of Biological, Biomedical, and Environmental Sciences, University of Hull, Cottingham Road, Hull HU6 7RX, UK; (I.J.F.D.); (R.I.L.M.)
- Correspondence: (J.M.R.); (J.C.); Tel.: +44-1482-465333 (J.M.R.); +852-3469-2124 (J.C.); Fax: +44-1482-465458 (J.M.R.); +852-3693-4766 (J.C.)
| | - Jinping Cheng
- State Key Laboratory of Estuarine and Coastal Research, East China Normal University, Shanghai 200062, China; (L.Q.); (Q.X.)
- Hong Kong Branch of Southern Marine Science and Engineering Guangdong Lab (Guangzhou) & Department of Ocean Science, School of Science, the Hong Kong University of Science and Technology, Kowloon, Hong Kong
- Correspondence: (J.M.R.); (J.C.); Tel.: +44-1482-465333 (J.M.R.); +852-3469-2124 (J.C.); Fax: +44-1482-465458 (J.M.R.); +852-3693-4766 (J.C.)
| |
Collapse
|
25
|
Flores EM, Nguyen AT, Odem MA, Eisenhoffer GT, Krachler AM. The zebrafish as a model for gastrointestinal tract-microbe interactions. Cell Microbiol 2020; 22:e13152. [PMID: 31872937 DOI: 10.1111/cmi.13152] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 11/07/2019] [Accepted: 12/19/2019] [Indexed: 02/06/2023]
Abstract
The zebrafish (Danio rerio) has become a widely used vertebrate model for bacterial, fungal, viral, and protozoan infections. Due to its genetic tractability, large clutch sizes, ease of manipulation, and optical transparency during early life stages, it is a particularly useful model to address questions about the cellular microbiology of host-microbe interactions. Although its use as a model for systemic infections, as well as infections localised to the hindbrain and swimbladder having been thoroughly reviewed, studies focusing on host-microbe interactions in the zebrafish gastrointestinal tract have been neglected. Here, we summarise recent findings regarding the developmental and immune biology of the gastrointestinal tract, drawing parallels to mammalian systems. We discuss the use of adult and larval zebrafish as models for gastrointestinal infections, and more generally, for studies of host-microbe interactions in the gut.
Collapse
Affiliation(s)
- Erika M Flores
- Department of Microbiology and Molecular Genetics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas.,M.D. Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, Texas
| | - Anh T Nguyen
- Department of Microbiology and Molecular Genetics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas.,M.D. Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, Texas
| | - Max A Odem
- Department of Microbiology and Molecular Genetics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas
| | - George T Eisenhoffer
- M.D. Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, Texas.,Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Anne Marie Krachler
- Department of Microbiology and Molecular Genetics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas.,M.D. Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, Texas
| |
Collapse
|
26
|
Flores-Cruz R, López-Arteaga R, Ramírez-Vidal L, López-Casillas F, Jiménez-Sánchez A. Unravelling the modus-operandi of chromenylium-cyanine fluorescent probes: a case study. Phys Chem Chem Phys 2019; 21:15779-15786. [PMID: 31282523 DOI: 10.1039/c9cp03256h] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Small-molecule fluorescent probes having optimized optical properties, such as high photostability and brightness, local microenvironment sensitivity and specific subcellular localizations, are increasingly available. Although the basis for designing efficient fluorophores for bioimaging applications is well established, implementing an improvement in a given photophysical characteristic always tends to compromise another optical property. This problem has enormous consequences for in vivo imaging, where ensuring a specific localization and precise control of the probe response is challenging. Herein we discuss a fluorescent probe, CC334, as a case study of the chromenylium-cyanine family that commonly exhibits highly complex photophysical schemes and highly interfered bioanalytical responses. By an exhaustive and concise analysis of the CC334 optical responses including detailed spectroscopic calibrations, steady-state microenvironment effects, ultrafast photophysics analysis and computational studies, we elucidate a new strategy to apply the probe in the singlet oxygen reactive oxygen species (1O2-ROS) monitoring using in vitro and in vivo models. The probe provides a new avenue for designing fluorescent probes to understand the dynamic behavior of subcellular environments.
Collapse
Affiliation(s)
- Ricardo Flores-Cruz
- Instituto de Química, Universidad Nacional Autónoma de México, México D.F., No. 04510, Mexico.
| | - Rafael López-Arteaga
- Instituto de Química, Universidad Nacional Autónoma de México, México D.F., No. 04510, Mexico.
| | - Lizbeth Ramírez-Vidal
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México D.F., No. 04510, Mexico
| | - Fernando López-Casillas
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México D.F., No. 04510, Mexico
| | - Arturo Jiménez-Sánchez
- Instituto de Química, Universidad Nacional Autónoma de México, México D.F., No. 04510, Mexico.
| |
Collapse
|
27
|
Etienne-Mesmin L, Chassaing B, Desvaux M, De Paepe K, Gresse R, Sauvaitre T, Forano E, de Wiele TV, Schüller S, Juge N, Blanquet-Diot S. Experimental models to study intestinal microbes–mucus interactions in health and disease. FEMS Microbiol Rev 2019; 43:457-489. [DOI: 10.1093/femsre/fuz013] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 05/31/2019] [Indexed: 02/06/2023] Open
Abstract
ABSTRACT
A close symbiotic relationship exists between the intestinal microbiota and its host. A critical component of gut homeostasis is the presence of a mucus layer covering the gastrointestinal tract. Mucus is a viscoelastic gel at the interface between the luminal content and the host tissue that provides a habitat to the gut microbiota and protects the intestinal epithelium. The review starts by setting up the biological context underpinning the need for experimental models to study gut bacteria-mucus interactions in the digestive environment. We provide an overview of the structure and function of intestinal mucus and mucins, their interactions with intestinal bacteria (including commensal, probiotics and pathogenic microorganisms) and their role in modulating health and disease states. We then describe the characteristics and potentials of experimental models currently available to study the mechanisms underpinning the interaction of mucus with gut microbes, including in vitro, ex vivo and in vivo models. We then discuss the limitations and challenges facing this field of research.
Collapse
Affiliation(s)
- Lucie Etienne-Mesmin
- Université Clermont Auvergne, INRA, MEDIS, 28 Place Henri Dunant, 63000 Clermont-Ferrand, France
| | - Benoit Chassaing
- Neuroscience Institute, Georgia State University, 100 Piedmont Ave SE, Atlanta, GA 30303 , USA
- Institute for Biomedical Sciences, Georgia State University, 100 Piedmont Ave, Atlanta, GA 30303 , USA
| | - Mickaël Desvaux
- Université Clermont Auvergne, INRA, MEDIS, 28 Place Henri Dunant, 63000 Clermont-Ferrand, France
| | - Kim De Paepe
- Center for Microbial Ecology and Technology (CMET), Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, 9000 Ghent, Belgium
| | - Raphaële Gresse
- Université Clermont Auvergne, INRA, MEDIS, 28 Place Henri Dunant, 63000 Clermont-Ferrand, France
| | - Thomas Sauvaitre
- Université Clermont Auvergne, INRA, MEDIS, 28 Place Henri Dunant, 63000 Clermont-Ferrand, France
| | - Evelyne Forano
- Université Clermont Auvergne, INRA, MEDIS, 28 Place Henri Dunant, 63000 Clermont-Ferrand, France
| | - Tom Van de Wiele
- Center for Microbial Ecology and Technology (CMET), Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, 9000 Ghent, Belgium
| | - Stephanie Schüller
- Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, United Kingdom
- Quadram Institute Bioscience, Norwich Research Park, Norwich NR7UQ, United Kingdom
| | - Nathalie Juge
- Quadram Institute Bioscience, Norwich Research Park, Norwich NR7UQ, United Kingdom
| | - Stéphanie Blanquet-Diot
- Université Clermont Auvergne, INRA, MEDIS, 28 Place Henri Dunant, 63000 Clermont-Ferrand, France
| |
Collapse
|
28
|
Cafora M, Deflorian G, Forti F, Ferrari L, Binelli G, Briani F, Ghisotti D, Pistocchi A. Phage therapy against Pseudomonas aeruginosa infections in a cystic fibrosis zebrafish model. Sci Rep 2019; 9:1527. [PMID: 30728389 PMCID: PMC6365511 DOI: 10.1038/s41598-018-37636-x] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 12/11/2018] [Indexed: 12/21/2022] Open
Abstract
Cystic fibrosis (CF) is a hereditary disease due to mutations in the CFTR gene and causes mortality in humans mainly due to respiratory infections caused by Pseudomonas aeruginosa. In a previous work we used phage therapy, which is a treatment with a mix of phages, to actively counteract acute P. aeruginosa infections in mice and Galleria mellonella larvae. In this work we apply phage therapy to the treatment of P. aeruginosa PAO1 infections in a CF zebrafish model. The structure of the CFTR channel is evolutionary conserved between fish and mammals and cftr-loss-of-function zebrafish embryos show a phenotype that recapitulates the human disease, in particular with destruction of the pancreas. We show that phage therapy is able to decrease lethality, bacterial burden, and the pro-inflammatory response caused by PAO1 infection. In addition, phage administration relieves the constitutive inflammatory state of CF embryos. To our knowledge, this is the first time that phage therapy is used to cure P. aeruginosa infections in a CF animal model. We also find that the curative effect against PAO1 infections is improved by combining phages and antibiotic treatments, opening a useful therapeutic approach that could reduce antibiotic doses and time of administration.
Collapse
Affiliation(s)
- Marco Cafora
- Dipartimento di Biotecnologie Mediche e Medicina Traslazionale, Università degli Studi di Milano - LITA, via Fratelli Cervi 93, 20090, Segrate, MI, Italy
| | - Gianluca Deflorian
- Istituto FIRC di Oncologia Molecolare - IFOM, Via Adamello 16, 20139, Milano, Italy
| | - Francesca Forti
- Dipartimento di Bioscienze, Università degli Studi di Milano, Via Celoria 26, 20133, Milano, Italy
| | - Laura Ferrari
- Istituto FIRC di Oncologia Molecolare - IFOM, Via Adamello 16, 20139, Milano, Italy
| | - Giorgio Binelli
- Dipartimento di Biotecnologie e Scienze della Vita, Università degli Studi dell'Insubria, Via J.H. Dunant 3, Varese, Italy
| | - Federica Briani
- Dipartimento di Bioscienze, Università degli Studi di Milano, Via Celoria 26, 20133, Milano, Italy
| | - Daniela Ghisotti
- Dipartimento di Bioscienze, Università degli Studi di Milano, Via Celoria 26, 20133, Milano, Italy
| | - Anna Pistocchi
- Dipartimento di Biotecnologie Mediche e Medicina Traslazionale, Università degli Studi di Milano - LITA, via Fratelli Cervi 93, 20090, Segrate, MI, Italy.
| |
Collapse
|
29
|
Depasquale JA. Actin Microridges. Anat Rec (Hoboken) 2018; 301:2037-2050. [DOI: 10.1002/ar.23965] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 05/03/2018] [Accepted: 05/14/2018] [Indexed: 12/21/2022]
|
30
|
DePasquale JA. Comparison of microridges in juvenile and adult sunfish,
Lepomis gibbosus. ACTA ZOOL-STOCKHOLM 2018. [DOI: 10.1111/azo.12281] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
31
|
Marcos-López M, Calduch-Giner JA, Mirimin L, MacCarthy E, Rodger HD, O'Connor I, Sitjà-Bobadilla A, Pérez-Sánchez J, Piazzon MC. Gene expression analysis of Atlantic salmon gills reveals mucin 5 and interleukin 4/13 as key molecules during amoebic gill disease. Sci Rep 2018; 8:13689. [PMID: 30209326 PMCID: PMC6135806 DOI: 10.1038/s41598-018-32019-8] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 08/20/2018] [Indexed: 12/19/2022] Open
Abstract
Amoebic gill disease (AGD) is one of the main diseases affecting Atlantic salmon (Salmo salar L.) mariculture. Hallmarks of AGD are hyperplasia of the lamellar epithelium and increased production of gill mucus. This study investigated the expression of genes involved in mucus secretion, cell cycle regulation, immunity and oxidative stress in gills using a targeted 21-gene PCR array. Gill samples were obtained from experimental and natural Neoparamoeba perurans infections, and sampling points included progressive infection stages and post-freshwater treatment. Up-regulation of genes related to mucin secretion and cell proliferation, and down-regulation of pro-inflammatory and pro-apoptotic genes were associated with AGD severity, while partial restoration of the gill homeostasis was detected post-treatment. Mucins and Th2 cytokines accoun ted for most of the variability observed between groups highlighting their key role in AGD. Two mucins (muc5, muc18) showed differential regulation upon disease. Substantial up-regulation of the secreted muc5 was detected in clinical AGD, and the membrane bound muc18 showed an opposite pattern. Th2 cytokines, il4/13a and il4/13b2, were significantly up-regulated from 2 days post-infection onwards, and changes were lesion-specific. Despite the differences between experimental and natural infections, both yielded comparable results that underline the importance of the studied genes in the respiratory organs of fish, and during AGD progression.
Collapse
Affiliation(s)
- Mar Marcos-López
- Marine and Freshwater Research Centre, Galway-Mayo Institute of Technology, Dublin Road, Galway, Co., Galway, H91 T8NW, Ireland. .,FishVet Group Ireland, Unit 7b Oranmore Business Park, Oranmore, Co, Galway, H91 XP3F, Ireland.
| | - Josep A Calduch-Giner
- Nutrigenomics and Fish Growth Endocrinology Group, Instituto de Acuicultura Torre de la Sal (IATS-CSIC), Castellón, 12595, Spain
| | - Luca Mirimin
- Marine and Freshwater Research Centre, Galway-Mayo Institute of Technology, Dublin Road, Galway, Co., Galway, H91 T8NW, Ireland
| | - Eugene MacCarthy
- Marine and Freshwater Research Centre, Galway-Mayo Institute of Technology, Dublin Road, Galway, Co., Galway, H91 T8NW, Ireland
| | - Hamish D Rodger
- FishVet Group Ireland, Unit 7b Oranmore Business Park, Oranmore, Co, Galway, H91 XP3F, Ireland
| | - Ian O'Connor
- Marine and Freshwater Research Centre, Galway-Mayo Institute of Technology, Dublin Road, Galway, Co., Galway, H91 T8NW, Ireland
| | - Ariadna Sitjà-Bobadilla
- Fish Pathology Group, Instituto de Acuicultura Torre de la Sal (IATS-CSIC), Castellón, 12595, Spain
| | - Jaume Pérez-Sánchez
- Nutrigenomics and Fish Growth Endocrinology Group, Instituto de Acuicultura Torre de la Sal (IATS-CSIC), Castellón, 12595, Spain
| | - M Carla Piazzon
- Fish Pathology Group, Instituto de Acuicultura Torre de la Sal (IATS-CSIC), Castellón, 12595, Spain.
| |
Collapse
|
32
|
Quintana-Hayashi MP, Padra M, Padra JT, Benktander J, Lindén SK. Mucus-Pathogen Interactions in the Gastrointestinal Tract of Farmed Animals. Microorganisms 2018; 6:E55. [PMID: 29912166 PMCID: PMC6027344 DOI: 10.3390/microorganisms6020055] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 06/09/2018] [Accepted: 06/15/2018] [Indexed: 12/29/2022] Open
Abstract
Gastrointestinal infections cause significant challenges and economic losses in animal husbandry. As pathogens becoming resistant to antibiotics are a growing concern worldwide, alternative strategies to treat infections in farmed animals are necessary in order to decrease the risk to human health and increase animal health and productivity. Mucosal surfaces are the most common route used by pathogens to enter the body. The mucosal surface that lines the gastrointestinal tract is covered by a continuously secreted mucus layer that protects the epithelial surface. The mucus layer is the first barrier the pathogen must overcome for successful colonization, and is mainly composed of densely glycosylated proteins called mucins. The vast array of carbohydrate structures present on the mucins provide an important setting for host-pathogen interactions. This review summarizes the current knowledge on gastrointestinal mucins and their role during infections in farmed animals. We examine the interactions between mucins and animal pathogens, with a focus on how pathogenic bacteria can modify the mucin environment in the gut, and how this in turn affects pathogen adhesion and growth. Finally, we discuss analytical challenges and complexities of the mucus-based defense, as well as its potential to control infections in farmed animals.
Collapse
Affiliation(s)
- Macarena P Quintana-Hayashi
- Department of Medical Biochemistry and Cell biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Box 440, 405 30 Gothenburg, Sweden.
| | - Médea Padra
- Department of Medical Biochemistry and Cell biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Box 440, 405 30 Gothenburg, Sweden.
| | - János Tamás Padra
- Department of Medical Biochemistry and Cell biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Box 440, 405 30 Gothenburg, Sweden.
| | - John Benktander
- Department of Medical Biochemistry and Cell biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Box 440, 405 30 Gothenburg, Sweden.
| | - Sara K Lindén
- Department of Medical Biochemistry and Cell biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Box 440, 405 30 Gothenburg, Sweden.
| |
Collapse
|
33
|
Bansil R, Turner BS. The biology of mucus: Composition, synthesis and organization. Adv Drug Deliv Rev 2018; 124:3-15. [PMID: 28970050 DOI: 10.1016/j.addr.2017.09.023] [Citation(s) in RCA: 408] [Impact Index Per Article: 58.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Revised: 09/24/2017] [Accepted: 09/27/2017] [Indexed: 02/06/2023]
Abstract
In this review we discuss mucus, the viscoelastic secretion from goblet or mucous producing cells that lines the epithelial surfaces of all organs exposed to the external world. Mucus is a complex aqueous fluid that owes its viscoelastic, lubricating and hydration properties to the glycoprotein mucin combined with electrolytes, lipids and other smaller proteins. Electron microscopy of mucosal surfaces reveals a highly convoluted surface with a network of fibers and pores of varying sizes. The major structural and functional component, mucin is a complex glycoprotein coded by about 20 mucin genes which produce a protein backbone having multiple tandem repeats of Serine, Threonine (ST repeats) where oligosaccharides are covalently O-linked. The N- and C-terminals of this apoprotein contain other domains with little or no glycosylation but rich in cysteines leading to dimerization and further multimerization via SS bonds. The synthesis of this complex protein starts in the endoplasmic reticulum with the formation of the apoprotein and is further modified via glycosylation in the cis and medial Golgi and packaged into mucin granules via Ca2+ bridging of the negative charges on the oligosaccharide brush in the trans Golgi. The mucin granules fuse with the plasma membrane of the secretory cells and following activation by signaling molecules release Ca2+ and undergo a dramatic change in volume due to hydration of the highly negatively charged polymer brush leading to exocytosis from the cells and forming the mucus layer. The rheological properties of mucus and its active component mucin and its mucoadhesivity are briefly discussed in light of their importance to mucosal drug delivery.
Collapse
|
34
|
Sveen LR, Grammes FT, Ytteborg E, Takle H, Jørgensen SM. Genome-wide analysis of Atlantic salmon (Salmo salar) mucin genes and their role as biomarkers. PLoS One 2017; 12:e0189103. [PMID: 29236729 PMCID: PMC5728529 DOI: 10.1371/journal.pone.0189103] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2017] [Accepted: 10/08/2017] [Indexed: 11/19/2022] Open
Abstract
The aim of this study was to identify potential mucin genes in the Atlantic salmon genome and evaluate tissue-specific distribution and transcriptional regulation in response to aquaculture-relevant stress conditions in post-smolts. Seven secreted gel-forming mucin genes were identified based on several layers of evidence; annotation, transcription, phylogeny and domain structure. Two genes were annotated as muc2 and five genes as muc5. The muc2 genes were predominantly transcribed in the intestinal region while the different genes in the muc5 family were mainly transcribed in either skin, gill or pyloric caeca. In order to investigate transcriptional regulation of mucins during stress conditions, two controlled experiments were conducted. In the first experiment, handling stress induced mucin transcription in the gill, while transcription decreased in the skin and intestine. In the second experiment, long term intensive rearing conditions (fish biomass ~125 kg/m3) interrupted by additional confinement led to increased transcription of mucin genes in the skin at one, seven and fourteen days post-confinement.
Collapse
Affiliation(s)
- Lene Rydal Sveen
- Department of Biology, Section of Marine Developmental Biology, University of Bergen (UiB), Bergen, Norway
- Division of Aquaculture, Section of Fish health, Norwegian Institute of Food, Fisheries and Aquaculture Research (Nofima), Ås, Norway
- * E-mail:
| | - Fabian Thomas Grammes
- Centre for Integrative Genetics (CIGENE), Department of Animal and Aquacultural Sciences (IHA), Faculty of Life Sciences (BIOVIT), Norwegian University of Life Sciences (NMBU), Ås, Norway
| | - Elisabeth Ytteborg
- Division of Aquaculture, Section of Fish health, Norwegian Institute of Food, Fisheries and Aquaculture Research (Nofima), Ås, Norway
| | - Harald Takle
- Division of Aquaculture, Section of Fish health, Norwegian Institute of Food, Fisheries and Aquaculture Research (Nofima), Ås, Norway
| | - Sven Martin Jørgensen
- Division of Aquaculture, Section of Fish health, Norwegian Institute of Food, Fisheries and Aquaculture Research (Nofima), Ås, Norway
| |
Collapse
|
35
|
Adamek M, Hazerli D, Matras M, Teitge F, Reichert M, Steinhagen D. Viral infections in common carp lead to a disturbance of mucin expression in mucosal tissues. FISH & SHELLFISH IMMUNOLOGY 2017; 71:353-358. [PMID: 29054826 DOI: 10.1016/j.fsi.2017.10.029] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 10/07/2017] [Accepted: 10/16/2017] [Indexed: 06/07/2023]
Abstract
In response to the constant challenge by potential pathogens, external surfaces of fish, their skin, gills and intestinal tract, are coated with mucus, a gel like substance which largely prevents the entry of pathogens. This mucus gel consists mainly of water and mucins, large O-glycosylated proteins, which are responsible for forming a gel like mixture. A modulation of the mRNA expression of mucins, was described in viral diseases in mammals however there is a knowledge gap about the regulation of mucins during viral infection in fish. Therefore, novel sequences for common carp mucins were located in an early version of the common carp genome and their mRNA expression measured in carp under infection with three different viral pathogens: (i) the alloherpesvirus cyprinid herpesvirus 3, (ii) the rhabdovirus spring viremia of carp virus and (iii) the poxvirus carp edema virus. The results showed a downregulation of mucin mRNA expression in gills and gut of common carp under infection with these pathogenic viruses. This could be a sign of a severe distress to the mucosal tissues in carp which occurs under viral infection. The reduced expression of mucins could help explaining the increased susceptibility of virus-infected carp to secondary bacterial infection.
Collapse
Affiliation(s)
- Mikolaj Adamek
- Fish Disease Research Unit, Institute for Parasitology, University of Veterinary Medicine, Bünteweg 17, 30559 Hannover, Germany.
| | - Dennis Hazerli
- Fish Disease Research Unit, Institute for Parasitology, University of Veterinary Medicine, Bünteweg 17, 30559 Hannover, Germany
| | - Marek Matras
- Laboratory of Fish Diseases, National Veterinary Research Institute, Partyzantów 57, 24-100 Puławy, Poland
| | - Felix Teitge
- Fish Disease Research Unit, Institute for Parasitology, University of Veterinary Medicine, Bünteweg 17, 30559 Hannover, Germany
| | - Michal Reichert
- Laboratory of Fish Diseases, National Veterinary Research Institute, Partyzantów 57, 24-100 Puławy, Poland
| | - Dieter Steinhagen
- Fish Disease Research Unit, Institute for Parasitology, University of Veterinary Medicine, Bünteweg 17, 30559 Hannover, Germany
| |
Collapse
|
36
|
Taormina MJ, Hay EA, Parthasarathy R. Passive and Active Microrheology of the Intestinal Fluid of the Larval Zebrafish. Biophys J 2017; 113:957-965. [PMID: 28834731 PMCID: PMC5567605 DOI: 10.1016/j.bpj.2017.06.069] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 06/12/2017] [Accepted: 06/26/2017] [Indexed: 12/24/2022] Open
Abstract
The fluids of the intestine serve as a physical barrier to pathogens, a medium for the diffusion of nutrients and metabolites, and an environment for commensal microbes. The rheological properties of intestinal mucus have therefore been the subject of many investigations, thus far limited to in vitro studies due to the difficulty of measurement in the natural context of the gut. This limitation especially hinders our understanding of how the gut microbiota interact with the intestinal space, since examination of this calls not only for in vivo measurement techniques, but for techniques that can be applied to model organisms in which the microbial state of the gut can be controlled. We have addressed this challenge with two complementary approaches. We performed passive microrheological measurements using thermally driven nanoparticles and active microrheology using micron-scale ellipsoidal magnetic microparticles, in both cases using light-sheet fluorescence microscopy to optically access the intestinal bulb of the larval zebrafish, a model vertebrate. We present viscosity measurements in germ-free animals (devoid of gut microbes), animals colonized by a single bacterial species, and conventionally reared animals, and find that in all cases, the mucin-rich intestinal liquid is well described as a Newtonian fluid. Surprisingly, despite known differences in the number of secretory cells in germ-free zebrafish and their conventional counterparts, the fluid viscosity for these two groups is very similar, as measured with either technique. Our study provides, to our knowledge, the first in vivo microrheological measurements of the intestinal space in living animals, and we comment on its implications for timescales of host-microbe interactions in the gut.
Collapse
Affiliation(s)
- Michael J Taormina
- Department of Physics, Institute of Molecular Biology, Materials Science Institute, University of Oregon, Eugene, Oregon
| | - Edouard A Hay
- Department of Physics, Institute of Molecular Biology, Materials Science Institute, University of Oregon, Eugene, Oregon
| | - Raghuveer Parthasarathy
- Department of Physics, Institute of Molecular Biology, Materials Science Institute, University of Oregon, Eugene, Oregon.
| |
Collapse
|
37
|
Portal C, Gouyer V, Magnien M, Plet S, Gottrand F, Desseyn JL. In vivo imaging of the Muc5b gel-forming mucin. Sci Rep 2017; 7:44591. [PMID: 28294161 PMCID: PMC5353722 DOI: 10.1038/srep44591] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 02/09/2017] [Indexed: 12/14/2022] Open
Abstract
Gel-forming mucins are macromolecules produced by goblet cells and responsible for the mucus gel formation. Changes in goblet cell density and in gel-forming mucin production have emerged as sensitive indicators for mucosal diseases. A Muc5b-GFP tagged reporter mouse was used to assess Muc5b production in mouse tissues by immunofluorescence microscopy and fluorescent activity using stereromicroscopy and probe-based confocal laser endomicroscopy. Muc5b production was followed longitudinally by recording the fluorescent activity in vagina and in embryonic lung explants under stimulation by interleukin 13. We show that the GFP is easily visualized in the mouse adult ear, nose, trachea, gallbladder, and cervix. Live Muc5b is also easily monitored in the nasal cavity, trachea and vagina where its production varies during the estrus cycle with a peak at the proestrus phase and in pregnant mice. Explant culture of reporter mouse embryonic whole lung shows that interleukin 13 stimulates Muc5b production. The transgenic Muc5b-GFP mouse is unique and suitable to study the mechanisms that regulate Muc5b production/secretion and mucous cell differentiation by live imaging and can be applied to test drug efficacy in mucosal disease models.
Collapse
Affiliation(s)
- Céline Portal
- LIRIC UMR 995, Univ. Lille, Inserm, CHU Lille, F-59000 Lille, France
| | - Valérie Gouyer
- LIRIC UMR 995, Univ. Lille, Inserm, CHU Lille, F-59000 Lille, France
| | - Mylène Magnien
- LIRIC UMR 995, Univ. Lille, Inserm, CHU Lille, F-59000 Lille, France
| | - Ségolène Plet
- LIRIC UMR 995, Univ. Lille, Inserm, CHU Lille, F-59000 Lille, France
| | - Frédéric Gottrand
- LIRIC UMR 995, Univ. Lille, Inserm, CHU Lille, F-59000 Lille, France
| | - Jean-Luc Desseyn
- LIRIC UMR 995, Univ. Lille, Inserm, CHU Lille, F-59000 Lille, France
| |
Collapse
|
38
|
De novo assembly of the sea trout (Salmo trutta m. trutta) skin transcriptome to identify putative genes involved in the immune response and epidermal mucus secretion. PLoS One 2017; 12:e0172282. [PMID: 28212382 PMCID: PMC5315281 DOI: 10.1371/journal.pone.0172282] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 02/02/2017] [Indexed: 01/02/2023] Open
Abstract
In fish, the skin is a multifunctional organ and the first barrier against pathogens. Salmonids differ in their susceptibility to microorganisms due to varied skin morphology and gene expression patterns. The brown trout is a salmonid species with important commercial and ecological value in Europe. However, there is a lack of knowledge regarding the genes involved in the immune response and mucus secretion in the skin of this fish. Thus, we characterized the skin transcriptome of anadromous brown trout using next-generation sequencing (NGS). A total of 1,348,306 filtered reads were obtained and assembled into 75,970 contigs. Of these contigs 48.57% were identified using BLAST tool searches against four public databases. KEGG pathway and Gene Ontology analyses revealed that 13.40% and 34.57% of the annotated transcripts, respectively, represent a variety of biological processes and functions. Among the identified KEGG Orthology categories, the best represented were signal transduction (23.28%) and immune system (8.82%), with a variety of genes involved in immune pathways, implying the differentiation of immune responses in the trout skin. We also identified and transcriptionally characterized 8 types of mucin proteins–the main structural components of the mucosal layer. Moreover, 140 genes involved in mucin synthesis were identified, and 1,119 potential simple sequence repeats (SSRs) were detected in 3,134 transcripts.
Collapse
|
39
|
Galindo-Villegas J, Garcia-Garcia E, Mulero V. Role of histamine in the regulation of intestinal immunity in fish. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2016; 64:178-186. [PMID: 26872545 DOI: 10.1016/j.dci.2016.02.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 02/02/2016] [Accepted: 02/08/2016] [Indexed: 06/05/2023]
Abstract
In mammals, during the acute inflammatory response, the complex interrelationship and cross-talk among histamine and the immune system has been fairly well characterized. There is a substantial body of information on its structure, metabolism, receptors, signal transduction, physiologic and pathologic effects. However, for early vertebrates, there is little such knowledge. In the case of teleost fish, this lack of knowledge has been due to the widely held belief that histamine is not present in this phylogenetic group. However, it has been recently demonstrated, that granules of mast cells in perciforms contain biologically active histamine. More importantly, the inflammatory response was clearly demonstrated to be regulated by the direct action of histamine on professional phagocytes. Nevertheless, the molecular basis and exact role of this biogenic amine in perciforms is still a matter of speculation. Therefore, this review intends to summarize recent experimental evidence regarding fish mast cells and correlate the same with their mammalian counterparts to establish the possible role of histamine in the fish intestinal inflammatory response.
Collapse
Affiliation(s)
- Jorge Galindo-Villegas
- Department of Cell Biology and Histology, Faculty of Biology, University of Murcia, IMIB-Arrixaca, Campus Universitario de Espinardo, Murcia 30100, Spain.
| | - Erick Garcia-Garcia
- Department of Cell Biology and Histology, Faculty of Biology, University of Murcia, IMIB-Arrixaca, Campus Universitario de Espinardo, Murcia 30100, Spain
| | - Victoriano Mulero
- Department of Cell Biology and Histology, Faculty of Biology, University of Murcia, IMIB-Arrixaca, Campus Universitario de Espinardo, Murcia 30100, Spain.
| |
Collapse
|
40
|
Eisenhoffer GT, Slattum G, Ruiz OE, Otsuna H, Bryan CD, Lopez J, Wagner DS, Bonkowsky JL, Chien CB, Dorsky RI, Rosenblatt J. A toolbox to study epidermal cell types in zebrafish. J Cell Sci 2016; 130:269-277. [PMID: 27149923 DOI: 10.1242/jcs.184341] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Accepted: 04/20/2016] [Indexed: 12/19/2022] Open
Abstract
Epithelia provide a crucial protective barrier for our organs and are also the sites where the majority of carcinomas form. Most studies on epithelia and carcinomas use cell culture or organisms where high-resolution live imaging is inaccessible without invasive techniques. Here, we introduce the developing zebrafish epidermis as an excellent in vivo model system for studying a living epithelium. We developed tools to fluorescently tag specific epithelial cell types and express genes in a mosaic fashion using five Gal4 lines identified from an enhancer trap screen. When crossed to a variety of UAS effector lines, we can now track, ablate or monitor single cells at sub-cellular resolution. Using photo-cleavable morpholino oligonucleotides that target gal4, we can also express genes in a mosaic fashion at specific times during development. Together, this system provides an excellent in vivo alternative to tissue culture cells, without the intrinsic concerns of culture conditions or transformation, and enables the investigation of distinct cell types within living epithelial tissues.
Collapse
Affiliation(s)
- George T Eisenhoffer
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Unit 1010, 1515 Holcombe Blvd., Houston, TX 77030-4009, USA
| | - Gloria Slattum
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, 2000 Circle of Hope Drive, Salt Lake City, UT 84112, USA
| | - Oscar E Ruiz
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Unit 1010, 1515 Holcombe Blvd., Houston, TX 77030-4009, USA
| | - Hideo Otsuna
- Department of Neurobiology and Anatomy, University of Utah School of Medicine, 320 BPRB, 20 South 2030 East, Salt Lake City, UT 84112, USA
| | - Chase D Bryan
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, 2000 Circle of Hope Drive, Salt Lake City, UT 84112, USA
| | - Justin Lopez
- Department of BioSciences, Rice University, W100 George R. Brown Hall, Houston, TX 77251-1892, USA
| | - Daniel S Wagner
- Department of BioSciences, Rice University, W100 George R. Brown Hall, Houston, TX 77251-1892, USA
| | - Joshua L Bonkowsky
- Department of Neurobiology and Anatomy, University of Utah School of Medicine, 320 BPRB, 20 South 2030 East, Salt Lake City, UT 84112, USA
| | - Chi-Bin Chien
- Department of Neurobiology and Anatomy, University of Utah School of Medicine, 320 BPRB, 20 South 2030 East, Salt Lake City, UT 84112, USA
| | - Richard I Dorsky
- Department of Neurobiology and Anatomy, University of Utah School of Medicine, 320 BPRB, 20 South 2030 East, Salt Lake City, UT 84112, USA
| | - Jody Rosenblatt
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, 2000 Circle of Hope Drive, Salt Lake City, UT 84112, USA
| |
Collapse
|
41
|
Lang T, Klasson S, Larsson E, Johansson MEV, Hansson GC, Samuelsson T. Searching the Evolutionary Origin of Epithelial Mucus Protein Components-Mucins and FCGBP. Mol Biol Evol 2016; 33:1921-36. [PMID: 27189557 PMCID: PMC4948705 DOI: 10.1093/molbev/msw066] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The gel-forming mucins are large glycosylated proteins that are essential components of the mucus layers covering epithelial cells. Using novel methods of identifying mucins based on profile hidden Markov models, we have found a large number of such proteins in Metazoa, aiding in their classification and allowing evolutionary studies. Most vertebrates have 5–6 gel-forming mucin genes and the genomic arrangement of these genes is well conserved throughout vertebrates. An exception is the frog Xenopus tropicalis with an expanded repertoire of at least 26 mucins of this type. Furthermore, we found that the ovomucin protein, originally identified in chicken, is characteristic of reptiles, birds, and amphibians. Muc6 is absent in teleost fish, but we now show that it is present in animals such as ghost sharks, demonstrating an early origin in vertebrate evolution. Public RNA-Seq data were analyzed with respect to mucins in zebrafish, frog, and chicken, thus allowing comparison in regard of tissue and developmental specificity. Analyses of invertebrate proteins reveal that gel-forming-mucin type of proteins is widely distributed also in this group. Their presence in Cnidaria, Porifera, and in Ctenophora (comb jellies) shows that these proteins were present early in metazoan evolution. Finally, we examined the evolution of the FCGBP protein, abundant in mucus and related to gel-forming mucins in terms of structure and localization. We demonstrate that FCGBP, ubiquitous in vertebrates, has a conserved N-terminal domain. Interestingly, this domain is also present as an N-terminal sequence in a number of bacterial proteins.
Collapse
Affiliation(s)
- Tiange Lang
- Department of Medical Biochemistry and Cell Biology, University of Gothenburg, Gothenburg, Sweden Key Laboratory of Tropical Plant Resource and Sustainable Use, Xishuangbanna Tropical Botanical Garden, Chinese Academy of Sciences, Mengla, Yunnan, China
| | - Sofia Klasson
- Department of Medical Biochemistry and Cell Biology, University of Gothenburg, Gothenburg, Sweden
| | - Erik Larsson
- Department of Medical Biochemistry and Cell Biology, University of Gothenburg, Gothenburg, Sweden
| | - Malin E V Johansson
- Department of Medical Biochemistry and Cell Biology, University of Gothenburg, Gothenburg, Sweden
| | - Gunnar C Hansson
- Department of Medical Biochemistry and Cell Biology, University of Gothenburg, Gothenburg, Sweden
| | - Tore Samuelsson
- Department of Medical Biochemistry and Cell Biology, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
42
|
|
43
|
Morales Fénero CI, Colombo Flores AA, Câmara NOS. Inflammatory diseases modelling in zebrafish. World J Exp Med 2016; 6:9-20. [PMID: 26929916 PMCID: PMC4759353 DOI: 10.5493/wjem.v6.i1.9] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Revised: 10/20/2015] [Accepted: 12/18/2015] [Indexed: 02/06/2023] Open
Abstract
The ingest of diets with high content of fats and carbohydrates, low or no physical exercise and a stressful routine are part of the everyday lifestyle of most people in the western world. These conditions are triggers for different diseases with complex interactions between the host genetics, the metabolism, the immune system and the microbiota, including inflammatory bowel diseases (IBD), obesity and diabetes. The incidence of these disorders is growing worldwide; therefore, new strategies for its study are needed. Nowadays, the majority of researches are in use of murine models for understand the genetics, physiopathology and interaction between cells and signaling pathways to find therapeutic solutions to these diseases. The zebrafish, a little tropical water fish, shares 70% of our genes and conserves anatomic and physiological characteristics, as well as metabolical pathways, with mammals, and is rising as a new complementary model for the study of metabolic and inflammatory diseases. Its high fecundity, fast development, transparency, versatility and low cost of maintenance makes the zebrafish an interesting option for new researches. In this review, we offer a discussion of the existing genetic and induced zebrafish models of two important Western diseases that have a strong inflammatory component, the IBD and the obesity.
Collapse
|
44
|
Protective and pro-inflammatory roles of intestinal bacteria. ACTA ACUST UNITED AC 2016; 23:67-80. [PMID: 26947707 DOI: 10.1016/j.pathophys.2016.02.002] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 02/11/2016] [Accepted: 02/12/2016] [Indexed: 02/06/2023]
Abstract
The intestinal mucosal surface in all vertebrates is exposed to enormous numbers of microorganisms that include bacteria, archaea, fungi and viruses. Coexistence of the host with the gut microbiota represents an active and mutually beneficial relationship that helps to shape the mucosal and systemic immune systems of both mammals and teleosts (ray-finned fish). Due to the potential for enteric microorganisms to invade intestinal tissue and induce local and/or systemic inflammation, the mucosal immune system has developed a number of protective mechanisms that allow the host to mount an appropriate immune response to invading bacteria, while limiting bystander tissue injury associated with these immune responses. Failure to properly regulate mucosal immunity is thought to be responsible for the development of chronic intestinal inflammation. The objective of this review is to present our current understanding of the role that intestinal bacteria play in vertebrate health and disease. While our primary focus will be humans and mice, we also present the new and exciting comparative studies being performed in zebrafish to model host-microbe interactions.
Collapse
|
45
|
Haarder S, Kania PW, Lindebo Holm T, Ohtani M, Buchmann K. Comparison of Two Chemically-Induced Colitis-Models in Adult Zebrafish, Using Optical Projection Tomography and Novel Transcriptional Markers. ACTA ACUST UNITED AC 2016. [DOI: 10.4236/oji.2016.64016] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
46
|
Jin C, Padra JT, Sundell K, Sundh H, Karlsson NG, Lindén SK. Atlantic Salmon Carries a Range of Novel O-Glycan Structures Differentially Localized on Skin and Intestinal Mucins. J Proteome Res 2015; 14:3239-51. [PMID: 26066491 DOI: 10.1021/acs.jproteome.5b00232] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Aquaculture is a growing industry, increasing the need for understanding host-pathogen interactions in fish. The skin and mucosal surfaces, covered by a mucus layer composed of mucins, is the first point of contact between fish and pathogens. Highly O-glycosylated mucins have been shown to be an important part of the defense against pathogens, and pathogens bind to host surfaces using lectin-like adhesins. However, knowledge of piscine O-glycosylation is very limited. We characterized mucin O-glycosylation of five freshwater acclimated Atlantic salmon, using mass spectrometry. Of the 109 O-glycans found, most were sialylated and differed in distribution among skin, pyloric ceca, and proximal and distal intestine. Skin O-glycans were shorter (2-6 residues) and less diverse (33 structures) than intestinal O-glycans (2-13 residues, 93 structures). Skin mucins carried O-glycan cores 1, 2, 3, and 5 and three types of sialic acids (Neu5Ac, Neu5Gc, and Kdn) and had sialyl-Tn as the predominant structure. Intestinal mucins carried only cores 1, 2, and 5, Neu5Ac was the only sialic acid present, and sialylated core 5 was the most dominant structure. This structural characterization can be used for identifying structures of putative importance in host-pathogen interactions for further testing in biological assays and disease intervention therapies.
Collapse
Affiliation(s)
- Chunsheng Jin
- †Department of Medical Chemistry and Cell Biology, ‡Department of Biological and Environmental Sciences, University of Gothenburg, Gothenburg SE-405 30, Sweden
| | - János Tamás Padra
- †Department of Medical Chemistry and Cell Biology, ‡Department of Biological and Environmental Sciences, University of Gothenburg, Gothenburg SE-405 30, Sweden
| | - Kristina Sundell
- †Department of Medical Chemistry and Cell Biology, ‡Department of Biological and Environmental Sciences, University of Gothenburg, Gothenburg SE-405 30, Sweden
| | - Henrik Sundh
- †Department of Medical Chemistry and Cell Biology, ‡Department of Biological and Environmental Sciences, University of Gothenburg, Gothenburg SE-405 30, Sweden
| | - Niclas G Karlsson
- †Department of Medical Chemistry and Cell Biology, ‡Department of Biological and Environmental Sciences, University of Gothenburg, Gothenburg SE-405 30, Sweden
| | - Sara K Lindén
- †Department of Medical Chemistry and Cell Biology, ‡Department of Biological and Environmental Sciences, University of Gothenburg, Gothenburg SE-405 30, Sweden
| |
Collapse
|