1
|
Madsen O, Rikkers RSC, Wells JM, Bergsma R, Kar SK, Taverne N, Taverne-Thiele AJ, Ellen ED, Woelders H. Transcriptomic analysis of intestinal organoids, derived from pigs divergent in feed efficiency, and their response to Escherichia coli. BMC Genomics 2024; 25:173. [PMID: 38350904 PMCID: PMC10863143 DOI: 10.1186/s12864-024-10064-0] [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: 05/10/2023] [Accepted: 01/30/2024] [Indexed: 02/15/2024] Open
Abstract
BACKGROUND There is increasing interest in using intestinal organoids to study complex traits like feed efficiency (FE) and host-microbe interactions. The aim of this study was to investigate differences in the molecular phenotype of organoids derived from pigs divergent for FE as well as their responses to challenge with adherent and invasive Escherichia coli (E. coli). RESULTS Colon and ileum tissue from low and high FE pigs was used to generate 3D organoids and two dimensional (2D) monolayers of organoid cells for E. coli challenge. Genome-wide gene expression was used to investigate molecular differences between pigs that were phenotypically divergent for FE and to study the difference in gene expression after challenge with E. coli. We showed, (1) minor differences in gene expression of colon organoids from pigs with low and high FE phenotypes, (2) that an E. coli challenge results in a strong innate immune gene response in both colon and ileum organoids, (3) that the immune response seems to be less pronounced in the colon organoids of high FE pigs and (4) a slightly stronger immune response was observed in ileum than in colon organoids. CONCLUSIONS These findings demonstrate the potential for using organoids to gain insights into complex biological mechanisms such as FE.
Collapse
Affiliation(s)
- Ole Madsen
- Animal Breeding & Genomics, Wageningen University & Research, PO Box 338, Wageningen, 6700 AH, the Netherlands.
| | - Roxann S C Rikkers
- Animal Breeding & Genomics, Wageningen University & Research, PO Box 338, Wageningen, 6700 AH, the Netherlands
| | - Jerry M Wells
- Host-Microbe Interactomics, Wageningen University & Research, PO Box 338, Wageningen, 6700 AH, the Netherlands
| | - Rob Bergsma
- Topigs Norsvin, Schoenaker 6, 6641 SZ, Beuningen, the Netherlands
| | - Soumya K Kar
- Animal Nutrition, Wageningen University & Research, PO Box 338, Wageningen, 6700 AH, the Netherlands
| | - Nico Taverne
- Host-Microbe Interactomics, Wageningen University & Research, PO Box 338, Wageningen, 6700 AH, the Netherlands
| | - Anja J Taverne-Thiele
- Host-Microbe Interactomics, Wageningen University & Research, PO Box 338, Wageningen, 6700 AH, the Netherlands
| | - Esther D Ellen
- Animal Breeding & Genomics, Wageningen University & Research, PO Box 338, Wageningen, 6700 AH, the Netherlands
| | - Henri Woelders
- Animal Breeding & Genomics, Wageningen University & Research, PO Box 338, Wageningen, 6700 AH, the Netherlands
| |
Collapse
|
2
|
Duarte ME, Garavito-Duarte Y, Kim SW. Impacts of F18 +Escherichia coli on Intestinal Health of Nursery Pigs and Dietary Interventions. Animals (Basel) 2023; 13:2791. [PMID: 37685055 PMCID: PMC10487041 DOI: 10.3390/ani13172791] [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: 08/15/2023] [Revised: 08/30/2023] [Accepted: 08/31/2023] [Indexed: 09/10/2023] Open
Abstract
This review focused on the impact of F18+E. coli on pig production and explored nutritional interventions to mitigate its deleterious effects. F18+E. coli is a primary cause of PWD in nursery pigs, resulting in substantial economic losses through diminished feed efficiency, morbidity, and mortality. In summary, the F18+E. coli induces intestinal inflammation with elevated IL6 (60%), IL8 (43%), and TNF-α (28%), disrupting the microbiota and resulting in 14% villus height reduction. Besides the mortality, the compromised intestinal health results in a 20% G:F decrease and a 10% ADFI reduction, ultimately culminating in a 28% ADG decrease. Among nutritional interventions to counter F18+E. coli impacts, zinc glycinate lowered TNF-α (26%) and protein carbonyl (45%) in jejunal mucosa, resulting in a 39% ADG increase. Lactic acid bacteria reduced TNF-α (36%), increasing 51% ADG, whereas Bacillus spp. reduced IL6 (27%), increasing BW (12%). Lactobacillus postbiotic increased BW (14%) and the diversity of beneficial bacteria. Phytobiotics reduced TNF-α (23%) and IL6 (21%), enhancing feed efficiency (37%). Additional interventions, including low crude protein formulation, antibacterial minerals, prebiotics, and organic acids, can be effectively used to combat F18+E. coli infection. These findings collectively underscore a range of effective strategies for managing the challenges posed by F18+E. coli in pig production.
Collapse
Affiliation(s)
| | | | - Sung Woo Kim
- Department of Animal Science, North Carolina State University, Raleigh, NC 27695, USA; (M.E.D.); (Y.G.-D.)
| |
Collapse
|
3
|
Ragan MV, Wala SJ, Sajankila N, Duff AF, Wang Y, Volpe SG, Al-Hadidi A, Dumbauld Z, Purayil N, Wickham J, Conces MR, Mihi B, Goodman SD, Bailey MT, Besner GE. Development of a novel definitive scoring system for an enteral feed-only model of necrotizing enterocolitis in piglets. Front Pediatr 2023; 11:1126552. [PMID: 37138566 PMCID: PMC10149862 DOI: 10.3389/fped.2023.1126552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Accepted: 03/28/2023] [Indexed: 05/05/2023] Open
Abstract
Introduction Necrotizing enterocolitis (NEC) is a complex inflammatory disorder of the human intestine that most often occurs in premature newborns. Animal models of NEC typically use mice or rats; however, pigs have emerged as a viable alternative given their similar size, intestinal development, and physiology compared to humans. While most piglet NEC models initially administer total parenteral nutrition prior to enteral feeds, here we describe an enteral-feed only piglet model of NEC that recapitulates the microbiome abnormalities present in neonates that develop NEC and introduce a novel multifactorial definitive NEC (D-NEC) scoring system to assess disease severity. Methods Premature piglets were delivered via Caesarean section. Piglets in the colostrum-fed group received bovine colostrum feeds only throughout the experiment. Piglets in the formula-fed group received colostrum for the first 24 h of life, followed by Neocate Junior to induce intestinal injury. The presence of at least 3 of the following 4 criteria were required to diagnose D-NEC: (1) gross injury score ≥4 of 6; (2) histologic injury score ≥3 of 5; (3) a newly developed clinical sickness score ≥5 of 8 within the last 12 h of life; and (4) bacterial translocation to ≥2 internal organs. Quantitative reverse transcription polymerase chain reaction was performed to confirm intestinal inflammation in the small intestine and colon. 16S rRNA sequencing was performed to evaluate the intestinal microbiome. Results Compared to the colostrum-fed group, the formula-fed group had lower survival, higher clinical sickness scores, and more severe gross and histologic intestinal injury. There was significantly increased bacterial translocation, D-NEC, and expression of IL-1α and IL-10 in the colon of formula-fed compared to colostrum-fed piglets. Intestinal microbiome analysis of piglets with D-NEC demonstrated lower microbial diversity and increased Gammaproteobacteria and Enterobacteriaceae. Conclusions We have developed a clinical sickness score and a new multifactorial D-NEC scoring system to accurately evaluate an enteral feed-only piglet model of NEC. Piglets with D-NEC had microbiome changes consistent with those seen in preterm infants with NEC. This model can be used to test future novel therapies to treat and prevent this devastating disease.
Collapse
Affiliation(s)
- Mecklin V. Ragan
- Center for Perinatal Research, Nationwide Children's Hospital, Columbus, OH, United States
- Department of Pediatric Surgery, Nationwide Children’s Hospital, Columbus, OH, United States
| | - Samantha J. Wala
- Center for Perinatal Research, Nationwide Children's Hospital, Columbus, OH, United States
- Department of Pediatric Surgery, Nationwide Children’s Hospital, Columbus, OH, United States
| | - Nitin Sajankila
- Center for Perinatal Research, Nationwide Children's Hospital, Columbus, OH, United States
- Department of Pediatric Surgery, Nationwide Children’s Hospital, Columbus, OH, United States
| | - Audrey F. Duff
- Center for Microbial Pathogenesis, Nationwide Children’s Hospital, Columbus, OH, United States
| | - Yijie Wang
- Center for Perinatal Research, Nationwide Children's Hospital, Columbus, OH, United States
| | - Samuel G. Volpe
- Center for Perinatal Research, Nationwide Children's Hospital, Columbus, OH, United States
| | - Ameer Al-Hadidi
- Center for Perinatal Research, Nationwide Children's Hospital, Columbus, OH, United States
| | - Zachary Dumbauld
- Center for Perinatal Research, Nationwide Children's Hospital, Columbus, OH, United States
| | - Nanditha Purayil
- Center for Perinatal Research, Nationwide Children's Hospital, Columbus, OH, United States
| | - Joseph Wickham
- Center for Microbial Pathogenesis, Nationwide Children’s Hospital, Columbus, OH, United States
| | - Miriam R. Conces
- Department of Pathology, Nationwide Children’s Hospital, Columbus, OH, United States
| | - Belgacem Mihi
- Center for Perinatal Research, Nationwide Children's Hospital, Columbus, OH, United States
| | - Steven D. Goodman
- Center for Microbial Pathogenesis, Nationwide Children’s Hospital, Columbus, OH, United States
| | - Michael T. Bailey
- Center for Microbial Pathogenesis, Nationwide Children’s Hospital, Columbus, OH, United States
| | - Gail E. Besner
- Center for Perinatal Research, Nationwide Children's Hospital, Columbus, OH, United States
- Department of Pediatric Surgery, Nationwide Children’s Hospital, Columbus, OH, United States
| |
Collapse
|
4
|
Wiarda JE, Becker SR, Sivasankaran SK, Loving CL. Regional epithelial cell diversity in the small intestine of pigs. J Anim Sci 2023; 101:skac318. [PMID: 36183288 PMCID: PMC9831138 DOI: 10.1093/jas/skac318] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 09/28/2022] [Indexed: 01/13/2023] Open
Abstract
Understanding regional distribution and specialization of small intestinal epithelial cells is crucial for developing methods to control appetite, stress, and nutrient uptake in swine. To establish a better understanding of specific epithelial cells found across different regions of the small intestine in pigs, we utilized single-cell RNA sequencing (scRNA-seq) to recover and analyze epithelial cells from duodenum, jejunum, and ileum. Cells identified included crypt cells, enterocytes, BEST4 enterocytes, goblet cells, and enteroendocrine (EE) cells. EE cells were divided into two subsets based on the level of expression of the EE lineage commitment gene, NEUROD1. NEUROD1hi EE cells had minimal expression of hormone-encoding genes and were dissimilar to EE cells in humans and mice, indicating a subset of EE cells unique to pigs. Recently discovered BEST4 enterocytes were detected in both crypts and villi throughout the small intestine via in situ staining, unlike in humans, where BEST4 enterocytes are found only in small intestinal villi. Proximal-to-distal gradients of expression were noted for hormone-encoding genes in EE cells and nutrient transport genes in enterocytes via scRNA-seq, demonstrating regional specialization. Regional gene expression in EE cells and enterocytes was validated via quantitative PCR (qPCR) analysis of RNA isolated from epithelial cells of different small intestinal locations. Though many genes had similar patterns of regional expression when assessed by qPCR of total epithelial cells, some regional expression was only detected via scRNA-seq, highlighting advantages of scRNA-seq to deconvolute cell type-specific regional gene expression when compared to analysis of bulk samples. Overall, results provide new information on regional localization and transcriptional profiles of epithelial cells in the pig small intestine.
Collapse
Affiliation(s)
- Jayne E Wiarda
- Food Safety and Enteric Pathogens Research Unit, National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Ames, IA, USA
- Immunobiology Graduate Program, Iowa State University, Ames, IA, USA
- Department of Veterinary Microbiology and Preventative Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA, USA
- Oak Ridge Institute for Science and Education, Agricultural Research Service Participation Program, Oak Ridge, TN, USA
| | - Sage R Becker
- Food Safety and Enteric Pathogens Research Unit, National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Ames, IA, USA
- Immunobiology Graduate Program, Iowa State University, Ames, IA, USA
- Department of Veterinary Microbiology and Preventative Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA, USA
| | - Sathesh K Sivasankaran
- Food Safety and Enteric Pathogens Research Unit, National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Ames, IA, USA
- Genome Informatics Facility, Iowa State University, Ames, IA, USA
| | - Crystal L Loving
- Food Safety and Enteric Pathogens Research Unit, National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Ames, IA, USA
- Immunobiology Graduate Program, Iowa State University, Ames, IA, USA
| |
Collapse
|
5
|
López-Martínez MJ, Cerón JJ, Ortín-Bustillo A, Escribano D, Kuleš J, Beletić A, Rubić I, González-Sánchez JC, Mrljak V, Martínez-Subiela S, Muñoz-Prieto A. A Proteomic Approach to Elucidate the Changes in Saliva and Serum Proteins of Pigs with Septic and Non-Septic Inflammation. Int J Mol Sci 2022; 23:ijms23126738. [PMID: 35743177 PMCID: PMC9223627 DOI: 10.3390/ijms23126738] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 06/10/2022] [Accepted: 06/15/2022] [Indexed: 12/29/2022] Open
Abstract
Sepsis is a systemic inflammatory response triggered by an infectious agent and is recognized by the World Health Organization as a global concern, since it is one of the major causes of severe illness in humans and animals. The study of the changes that can occur in saliva and serum in sepsis can contribute to a better understanding of the pathophysiological mechanisms involved in the process and also to discover potential biomarkers that can help in its diagnosis and monitoring. The objective of this study was to characterize the changes that occur in the salivary and serum proteome of pigs with experimentally-induced sepsis. The study included five pigs with sepsis induced by LPS administration and five pigs with non-septic inflammation induced by turpentine for comparative purposes. In saliva, there were eighteen salivary proteins differentially expressed in the sepsis condition and nine in non-septic inflammation. Among these, significant increments in aldolase A and serpin B12 only occurred in the sepsis model. Changes in aldolase A were validated in a larger population of pigs with sepsis due to Streptococcus suis infection. In serum, there were 30 proteins differentially expressed in sepsis group and 26 proteins in the non-septic group, and most of the proteins that changed in both groups were related to non-specific inflammation. In the saliva of the septic animals there were some specific pathways activated, such as the organonitrogen compound metabolic process and lipid transport, whereas, in the serum, one of the main activated pathways was the regulation of protein secretion. Overall, saliva and serum showed different proteome variations in response to septic inflammation and could provide complementary information about the pathophysiological mechanisms occurring in this condition. Additionally, salivary aldolase A could be a potential biomarker of sepsis in pigs that should be confirmed in a larger population.
Collapse
Affiliation(s)
- María José López-Martínez
- Interdisciplinary Laboratory of Clinical Analysis of the University of Murcia (Interlab-UMU), Department of Animal Medicine and Surgery, Veterinary School, Regional Campus of International Excellence Mare Nostrum, University of Murcia, Espinardo, 30100 Murcia, Spain; (M.J.L.-M.); (A.O.-B.); (D.E.); (I.R.); (S.M.-S.)
| | - José Joaquín Cerón
- Interdisciplinary Laboratory of Clinical Analysis of the University of Murcia (Interlab-UMU), Department of Animal Medicine and Surgery, Veterinary School, Regional Campus of International Excellence Mare Nostrum, University of Murcia, Espinardo, 30100 Murcia, Spain; (M.J.L.-M.); (A.O.-B.); (D.E.); (I.R.); (S.M.-S.)
- Correspondence: (J.J.C.); (A.M.-P.)
| | - Alba Ortín-Bustillo
- Interdisciplinary Laboratory of Clinical Analysis of the University of Murcia (Interlab-UMU), Department of Animal Medicine and Surgery, Veterinary School, Regional Campus of International Excellence Mare Nostrum, University of Murcia, Espinardo, 30100 Murcia, Spain; (M.J.L.-M.); (A.O.-B.); (D.E.); (I.R.); (S.M.-S.)
| | - Damián Escribano
- Interdisciplinary Laboratory of Clinical Analysis of the University of Murcia (Interlab-UMU), Department of Animal Medicine and Surgery, Veterinary School, Regional Campus of International Excellence Mare Nostrum, University of Murcia, Espinardo, 30100 Murcia, Spain; (M.J.L.-M.); (A.O.-B.); (D.E.); (I.R.); (S.M.-S.)
| | - Josipa Kuleš
- Clinic for Internal Diseases, Faculty of Veterinary Medicine, University of Zagreb, Heinzelova 55, 10000 Zagreb, Croatia; (J.K.); (A.B.); (V.M.)
| | - Anđelo Beletić
- Clinic for Internal Diseases, Faculty of Veterinary Medicine, University of Zagreb, Heinzelova 55, 10000 Zagreb, Croatia; (J.K.); (A.B.); (V.M.)
| | - Ivana Rubić
- Interdisciplinary Laboratory of Clinical Analysis of the University of Murcia (Interlab-UMU), Department of Animal Medicine and Surgery, Veterinary School, Regional Campus of International Excellence Mare Nostrum, University of Murcia, Espinardo, 30100 Murcia, Spain; (M.J.L.-M.); (A.O.-B.); (D.E.); (I.R.); (S.M.-S.)
| | | | - Vladimir Mrljak
- Clinic for Internal Diseases, Faculty of Veterinary Medicine, University of Zagreb, Heinzelova 55, 10000 Zagreb, Croatia; (J.K.); (A.B.); (V.M.)
| | - Silvia Martínez-Subiela
- Interdisciplinary Laboratory of Clinical Analysis of the University of Murcia (Interlab-UMU), Department of Animal Medicine and Surgery, Veterinary School, Regional Campus of International Excellence Mare Nostrum, University of Murcia, Espinardo, 30100 Murcia, Spain; (M.J.L.-M.); (A.O.-B.); (D.E.); (I.R.); (S.M.-S.)
| | - Alberto Muñoz-Prieto
- Interdisciplinary Laboratory of Clinical Analysis of the University of Murcia (Interlab-UMU), Department of Animal Medicine and Surgery, Veterinary School, Regional Campus of International Excellence Mare Nostrum, University of Murcia, Espinardo, 30100 Murcia, Spain; (M.J.L.-M.); (A.O.-B.); (D.E.); (I.R.); (S.M.-S.)
- Clinic for Internal Diseases, Faculty of Veterinary Medicine, University of Zagreb, Heinzelova 55, 10000 Zagreb, Croatia; (J.K.); (A.B.); (V.M.)
- Correspondence: (J.J.C.); (A.M.-P.)
| |
Collapse
|
6
|
Alves L, de Novais FJ, da Silva AN, Araujo MS, Bernardino T, Osowski GV, Zanella R, Lee Settles M, Holmes MA, Fukumasu H, Ruiz VLDA, Zanella AJ. Vaginal Microbiota Diversity in Response to Lipopolysaccharide in Gilts Housed Under Three Housing Systems. Front Genet 2022; 13:836962. [PMID: 35464863 PMCID: PMC9024362 DOI: 10.3389/fgene.2022.836962] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 03/04/2022] [Indexed: 12/13/2022] Open
Abstract
The United Kingdom and European Union have banned crates for pregnant sows. However, animals are kept in a restrictive environment for up to four weeks after mating, leading to stress and different responses of the animals’ immune system. Here, we used vaginal flushing of gilts to investigate whether housing systems or an experimental inflammatory challenge with lipopolysaccharide (LPS) can modify the gilt vaginal microbiome. Alpha-diversity indices showed differences in the microbiota of gilts housed under different systems (q = 0.04). Shannon alpha-diversity richness was higher in gilts group-housed in pens than in gilts housed in crates (q = 0.035), but not higher than in other groups. The relative abundance of the operational taxonomic unit (OTU) (q < 0.05) revealed specific differences in housing systems before a LPS or saline (SAL control) challenge. We found different abundances in taxa of Actinobacteria, Bacteroidetes, Cyanobacteria, Firmicutes, and Proteobacteria in gilts housed in the different systems before challenge. After the LPS challenge, significant differences were detected in the relative abundance of OTUs (q < 0.05) for the LPS-challenged group compared with SAL animals for each housing system. The phylum Staphylococcus showed higher abundance among the LPS-challenged gilts than in SAL-challenged animals. Furthermore, Enterobacter was more abundant in the LPS-challenged gilts housed in crates than in SAL-challenged gilts housed in crates. Streptococcus suis, Conchiformibius, Globicatella and Actinobacillus were more abundant in LPS-challenged gilts in indoor group housing than in SAL gilts in the same housing system. Gilts kept outdoors did not show changes in vaginal microbiota after an LPS challenge. Gilts housed in crates showed clinical signs of urogenital infection, whereas gilts housed outdoors and in indoor group housing did not. The relationship between environment, immune response, and microbiota suggested that animals in a poor environments experience difficulties responding to a challenge and their vaginal microbiota is altered as a consequence, with decreased richness of normal vaginal microbiota, and increased opportunistic bacteria. Welfare indicators measured by gilts’ responses to housing systems however, do not fully explain mechanisms associated with the unique signature in vaginal microbiota encountered in the different housing systems.
Collapse
Affiliation(s)
- Luana Alves
- Department of Veterinary Medicine and Animal Health, Faculty of Veterinary Medicine and Animal Science, University of São Paulo, Pirassununga, Brazil
| | - Francisco José de Novais
- Department of Veterinary Medicine, Faculty of Animal Science and Food Engineering, University of São Paulo, Pirassununga, Brazil
| | - Arthur Nery da Silva
- Department of Veterinary Medicine and Animal Health, Faculty of Veterinary Medicine and Animal Science, University of São Paulo, Pirassununga, Brazil
| | - Michelle Silva Araujo
- Department of Veterinary Medicine and Animal Health, Faculty of Veterinary Medicine and Animal Science, University of São Paulo, Pirassununga, Brazil
| | - Thiago Bernardino
- Department of Veterinary Medicine and Animal Health, Faculty of Veterinary Medicine and Animal Science, University of São Paulo, Pirassununga, Brazil.,Graduation Program in One Health, University of Santo Amaro, São Paulo, Brazil
| | - Germana Vizzotto Osowski
- Department of Veterinary Medicine and Animal Health, Faculty of Veterinary Medicine and Animal Science, University of São Paulo, Pirassununga, Brazil
| | - Ricardo Zanella
- Faculty of Agronomy and Veterinary Medicine, University of Passo Fundo, Passo Fundo, Brazil
| | - Matthew Lee Settles
- Director of Bioinformatics Core, UC Davis Genome Center, Davis, CA, United States
| | - Mark A Holmes
- Department of Veterinary Medicine, School of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Heidge Fukumasu
- Department of Veterinary Medicine, Faculty of Animal Science and Food Engineering, University of São Paulo, Pirassununga, Brazil
| | - Vera Letticie de Azevedo Ruiz
- Department of Veterinary Medicine, Faculty of Animal Science and Food Engineering, University of São Paulo, Pirassununga, Brazil
| | - Adroaldo José Zanella
- Department of Veterinary Medicine and Animal Health, Faculty of Veterinary Medicine and Animal Science, University of São Paulo, Pirassununga, Brazil
| |
Collapse
|
7
|
Zhong T, Feng M, Su M, Wang D, Li Q, Jia S, Luo F, Wang H, Hu E, Yang X, Fan Y. Qihuzha granule attenuated LPS-induced acute spleen injury in mice via Src/MAPK/Stat3 signal pathway. JOURNAL OF ETHNOPHARMACOLOGY 2021; 281:114458. [PMID: 34352329 DOI: 10.1016/j.jep.2021.114458] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 07/13/2021] [Accepted: 07/24/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Qihuzha granule (QHZG), is one of traditional Chinese patent medicines composed of eleven edible medicinal plant, which has been used in the clinic for the treatment of indigestion and anorexia in children caused by deficiency of the spleen and stomach. Yet it is noteworthy that QHZG has therapeutic effect on recurrent respiratory tract infection (RRTI) in children. However, its potential molecular mechanisms remained unclear. AIM OF THE STUDY The aim of this study was to investigate the therapeutic effect and potential mechanism of QHZG on lipopolysaccharide (LPS) induced acute spleen injury. MATERIALS AND METHODS The acute spleen injury model was induced by intraperitoneal injection of LPS (10 mg/kg) and safe doses of QHZG was administered by gavage once a day for 23 days before LPS treatment. Serum inflammatory cytokines including interleukin-2 (IL-2), IL-1β, IFN-γ, and tumor necrosis factor-α (TNF-α) were tested by ELISA. Related protein levels were detected by Western blotting. Hematoxylin-eosin (HE) staining was employed to observe the histological alterations. The distribution of macrophages and neutrophils in the mouse spleen was examined by immunofluorescence analysis. RESULTS QHZG pretreatment significantly abolished the increased secretion of cytokines such as interleukin-2 (IL-2), IL-1β, IFN-γ, and tumor necrosis factor-α (TNF-α), which were attributable to LPS treatment. Immunofluorescence staining and Histological analysis of spleen tissue revealed the protective effect of QHZG against LPS-induced acute spleen injury in mice. Further study indicated that pretreatment with QHZG significantly inhibited LPS-induced phosphorylation of Src. Accordingly, the increased phosphorylation of Src downstream components (JNK, ERK, P38 and STAT3) induced by LPS was remarkably diminished by QHZG, suggesting the involvement of Src/MAPK/STAT3 pathway in the inhibitory effects of QHZG on spleen injury in mice. CONCLUSION Our study demonstrated that QHZG protected mice from LPS-induced acute spleen injury via inhibition of Src/MAPK/Stat3 signal pathway. These results suggested that QHZG might serve as a new drug for the treatment of LPS-stimulated spleen injury.
Collapse
Affiliation(s)
- Ting Zhong
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, PR China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang, 550014, PR China
| | - Min Feng
- Sunflower Pharmaceutical Group (Guizhou) Hongqi Co., Ltd, Liupanshui, 553400, PR China
| | - Minzhi Su
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, PR China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang, 550014, PR China
| | - Daoping Wang
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, PR China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang, 550014, PR China
| | - Qing Li
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, PR China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang, 550014, PR China
| | - Shuqin Jia
- Guiyang First People's Hospital, Guiyang, 550014, PR China
| | - Fang Luo
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, PR China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang, 550014, PR China
| | - Haibo Wang
- Sunflower Pharmaceutical Group (Guizhou) Hongqi Co., Ltd, Liupanshui, 553400, PR China
| | - Enming Hu
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, PR China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang, 550014, PR China
| | - Xiaosheng Yang
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, PR China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang, 550014, PR China.
| | - Yanhua Fan
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, PR China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang, 550014, PR China.
| |
Collapse
|
8
|
Zhang Y, Duan X, Wassie T, Wang HH, Li T, Xie C, Wu X. Enteromorpha prolifera polysaccharide-zinc complex modulates the immune response and alleviates LPS-induced intestinal inflammation via inhibiting the TLR4/NF-κB signaling pathway. Food Funct 2021; 13:52-63. [PMID: 34704575 DOI: 10.1039/d1fo02171k] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Enteromorpha prolifera polysaccharide-zinc (EP-Zn), a kind of polysaccharide-zinc complex, has been shown to improve the immune response and reduce the inflammatory factors in weaned piglets. Yet, the molecular mechanism remains unclear. The present study was conducted to investigate the immunomodulating activity and anti-inflammatory mechanism of EP-Zn in mice. Different doses (350 mg kg-1, 700 mg kg-1, 1050 mg kg-1 and 1400 mg kg-1) of EP-Zn were administered to C57BL/6J mice for 28 days. The results showed that under physiological conditions, 350 mg kg-1 EP-Zn stimulated cytokine (TNF-α, IL-1β, IL-6 and IL-10) secrection, regulated the intestinal microbiota, and reduced the levels of short-chain fatty acids (SCFAs) (acetic acid and propionic acid). In addition, in the LPS-induced inflammation model, EP-Zn pretreatment effectively alleviated LPS-induced shortening of colonic length and increased MPO and DAO contents, improved intestinal physical barrier function by modulating mucosal structure, and attenuated intestinal inflammation via inhibiting the TLR4/NF-κB signaling pathway. These findings suggested that EP-Zn exerted immunomodulatory and anti-inflammatory activities under physiological and inflammatory conditions, respectively.
Collapse
Affiliation(s)
- Yumei Zhang
- College of Resources and Environment, College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, Hunan 410128, China. .,CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences; National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Changsha, Hunan 410125, China
| | - Xinyi Duan
- College of Resources and Environment, College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, Hunan 410128, China. .,CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences; National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Changsha, Hunan 410125, China
| | - Teketay Wassie
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences; National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Changsha, Hunan 410125, China
| | - Hai-Hua Wang
- Qingdao Seawin Biotech Group Co., Ltd., Qingdao, 266071, China
| | - Tiejun Li
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences; National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Changsha, Hunan 410125, China
| | - Chunyan Xie
- College of Resources and Environment, College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, Hunan 410128, China.
| | - Xin Wu
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences; National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Changsha, Hunan 410125, China.,Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, 300308, P R China
| |
Collapse
|
9
|
Messad F, Louveau I, Renaudeau D, Gilbert H, Gondret F. Analysis of merged whole blood transcriptomic datasets to identify circulating molecular biomarkers of feed efficiency in growing pigs. BMC Genomics 2021; 22:501. [PMID: 34217223 PMCID: PMC8254903 DOI: 10.1186/s12864-021-07843-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 06/24/2021] [Indexed: 11/10/2022] Open
Abstract
Background Improving feed efficiency (FE) is an important goal due to its economic and environmental significance for farm animal production. The FE phenotype is complex and based on the measurements of the individual feed consumption and average daily gain during a test period, which is costly and time-consuming. The identification of reliable predictors of FE is a strategy to reduce phenotyping efforts. Results Gene expression data of the whole blood from three independent experiments were combined and analyzed by machine learning algorithms to propose molecular biomarkers of FE traits in growing pigs. These datasets included Large White pigs from two lines divergently selected for residual feed intake (RFI), a measure of net FE, and in which individual feed conversion ratio (FCR) and blood microarray data were available. Merging the three datasets allowed considering FCR values (Mean = 2.85; Min = 1.92; Max = 5.00) for a total of n = 148 pigs, with a large range of body weight (15 to 115 kg) and different test period duration (2 to 9 weeks). Random forest (RF) and gradient tree boosting (GTB) were applied on the whole blood transcripts (26,687 annotated molecular probes) to identify the most important variables for binary classification on RFI groups and a quantitative prediction of FCR, respectively. The dataset was split into learning (n = 74) and validation sets (n = 74). With iterative steps for variable selection, about three hundred’s (328 to 391) molecular probes participating in various biological pathways, were identified as important predictors of RFI or FCR. With the GTB algorithm, simpler models were proposed combining 34 expressed unique genes to classify pigs into RFI groups (100% of success), and 25 expressed unique genes to predict FCR values (R2 = 0.80, RMSE = 8%). The accuracy performance of RF models was slightly lower in classification and markedly lower in regression. Conclusion From small subsets of genes expressed in the whole blood, it is possible to predict the binary class and the individual value of feed efficiency. These predictive models offer good perspectives to identify animals with higher feed efficiency in precision farming applications. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-021-07843-4.
Collapse
Affiliation(s)
- Farouk Messad
- PEGASE, INRAE, Institut Agro, 35590, Saint-Gilles, France
| | | | | | - Hélène Gilbert
- GenPhySE, INRAE, INP-ENVT, 31326, Castanet Tolosan, France
| | | |
Collapse
|
10
|
Käser T. Swine as biomedical animal model for T-cell research-Success and potential for transmittable and non-transmittable human diseases. Mol Immunol 2021; 135:95-115. [PMID: 33873098 DOI: 10.1016/j.molimm.2021.04.004] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 03/23/2021] [Accepted: 04/01/2021] [Indexed: 02/07/2023]
Abstract
Swine is biologically one of the most relevant large animal models for biomedical research. With its use as food animal that can be exploited as a free cell and tissue source for research and its high susceptibility to human diseases, swine additionally represent an excellent option for both the 3R principle and One Health research. One of the previously most limiting factors of the pig model was its arguably limited immunological toolbox. Yet, in the last decade, this toolbox has vastly improved including the ability to study porcine T-cells. This review summarizes the swine model for biomedical research with focus on T cells. It first contrasts the swine model to the more commonly used mouse and non-human primate model before describing the current capabilities to characterize and extend our knowledge on porcine T cells. Thereafter, it not only reflects on previous biomedical T-cell research but also extends into areas in which more in-depth T-cell analyses could strongly benefit biomedical research. While the former should inform on the successes of biomedical T-cell research in swine, the latter shall inspire swine T-cell researchers to find collaborations with researchers working in other areas - such as nutrition, allergy, cancer, transplantation, infectious diseases, or vaccine development.
Collapse
Affiliation(s)
- Tobias Käser
- Department of Population Health and Pathobiology, College of Veterinary Medicine, North Carolina State University, 1060 William Moore Drive, 27607 Raleigh, NC, USA.
| |
Collapse
|