1
|
Liang J, Wang S, Kou S, Chen C, Zhang W, Nie C. Clostridium butyricum Prevents Diarrhea Incidence in Weaned Piglets Induced by Escherichia coli K88 through Rectal Bacteria-Host Metabolic Cross-Talk. Animals (Basel) 2024; 14:2287. [PMID: 39199821 PMCID: PMC11350811 DOI: 10.3390/ani14162287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 07/15/2024] [Accepted: 07/30/2024] [Indexed: 09/01/2024] Open
Abstract
This study aimed to evaluate the effects of Clostridium butyricum (C. butyricum) on the prevention of the diarrhea rates and growth performances of weaned piglets induced by Escherichia coli K88 (E. coli K88). Twenty-four weaned piglets (6.92 ± 0.11 kg) were randomly assigned to one of three treatment groups for a period of 21 days. Each group consisted of eight pigs, with each pig being housed in an individual pen. Group I received the control diet along with normal saline, Group II received the control diet along with E. coli K88, and Group III received the control diet supplemented with 5 × 108 CFU/kg of C. butyricum and E. coli K88. We examined alterations in rectal microbiota and metabolites, analyzed the incidence of diarrhea, and investigated the interactions between microbiota and metabolites through the application of Illumina MiSeq sequencing and liquid chromatography-mass spectrometry. The results showed that, from days 14 to 21, the diarrhea incidence in Group III decreased significantly by 83.29% compared to Group II (p < 0.05). Over the entire experimental duration, the average daily feed intake of Group III decreased significantly by 11.13% compared to Group I (p < 0.05), while the diarrhea incidence in Group III decreased by 71.46% compared to Group II (p < 0.05). The predominant microbial flora in the rectum consisted of Firmicutes (57.32%), Bacteroidetes (41.03%), and Proteobacteria (0.66%). Administering E. coli K88 orally can elevate the relative abundance of Megasphaera (p < 0.05). Conversely, the supplementation of C. butyricum in the diet reduced the relative abundance of Megasphaera (p < 0.05), while increasing the relative abundance of unclassified_f_Lachnospiraceae (p < 0.05). Rectal metabolomics analysis revealed that supplementing C. butyricum in the feed significantly altered the amino acids and fatty acids of the piglets infected with E. coli K88 (p < 0.05). The correlation analysis showed that the occurrence of diarrhea was inversely related to adipic acid (p < 0.05) and positively associated with (5-hydroxyindol-3-YL) acetic acid and L-aspartic acid (p < 0.05). Prevotella_1 exhibited a negative correlation with octadecanoic acid (p < 0.05). Prevotellaceae_UCG-005 showed a negative correlation with (5-hydroxyindol-3-YL) acetic acid (p < 0.05). The findings from this research study aid in probiotic development and the enhancement of healthy growth in weaned piglets.
Collapse
Affiliation(s)
- Jing Liang
- College of Life Science, Yulin University, Yulin 719000, China; (J.L.); (S.W.)
- College of Animal Science and Technology, Shihezi University, Shihezi 832003, China; (S.K.); (C.C.)
| | - Sihu Wang
- College of Life Science, Yulin University, Yulin 719000, China; (J.L.); (S.W.)
- College of Animal Science and Technology, Northwest A&F University, Xianyang 712100, China
| | - Shasha Kou
- College of Animal Science and Technology, Shihezi University, Shihezi 832003, China; (S.K.); (C.C.)
| | - Cheng Chen
- College of Animal Science and Technology, Shihezi University, Shihezi 832003, China; (S.K.); (C.C.)
| | - Wenju Zhang
- College of Animal Science and Technology, Shihezi University, Shihezi 832003, China; (S.K.); (C.C.)
| | - Cunxi Nie
- College of Animal Science and Technology, Shihezi University, Shihezi 832003, China; (S.K.); (C.C.)
| |
Collapse
|
2
|
Vega-Munguía G, Vargas Sánchez A, Camacho-Medina JE, Suárez-Vélez L, Bárcenas-Morales G, Quintar Guerrero D, Ciprian-Carrasco A, Mendoza Elvira S. Effect of Live and Fragmented Saccharomyces cerevisiae in the Feed of Pigs Challenged with Mycoplasma hyopneumoniae. Pathogens 2024; 13:322. [PMID: 38668277 PMCID: PMC11054539 DOI: 10.3390/pathogens13040322] [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: 02/16/2024] [Revised: 03/22/2024] [Accepted: 04/05/2024] [Indexed: 04/29/2024] Open
Abstract
Currently, the responsible use of antimicrobials in pigs has allowed the continuous development of alternatives to these antimicrobials. In this study, we describe the impact of treatments with two probiotics, one based on live Saccharomyces cerevisiae (S. cerevisiae) and another based on fragmented S. cerevisiae (beta-glucans), that were administered to piglets at birth and at prechallenge with Mycoplasma hyopneumoniae. Thirty-two pigs were divided into four groups of eight animals each. The animals had free access to water and food. The groups were as follows: Group A, untreated negative control; Group B, inoculated by nebulization with M. hyopneumoniae positive control; Group C, first treated with disintegrated S. cerevisiae (disintegrated Sc) and inoculated by nebulization with M. hyopneumoniae; and Group D, treated with live S. cerevisiae yeast (live Sc) and inoculated by nebulization with M. hyopneumoniae. In a previous study, we found that on Days 1 and 21 of blood sampling, nine proinflammatory cytokines were secreted, and an increase in their secretion occurred for only five of them: TNF-α, INF-α, INF-γ, IL-10, and IL-12 p40. The results of the clinical evolution, the degree of pneumonic lesions, and the productive parameters of treated Groups C and D suggest that S. cerevisiae has an immunomodulatory effect in chronic proliferative M. hyopneumoniae pneumonia characterized by delayed hypersensitivity, which depends on the alteration or modulation of the respiratory immune response. The data presented in this study showed that S. cerevisiae contributed to the innate resistance of infected pigs.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Susana Mendoza Elvira
- Department of Biological Sciences, Facultad de Estudios Superiores Cuatitlan, Universidad Nacional Autónoma de México, Cuautitlán Izcalli 54720, Mexico; (G.V.-M.); (J.E.C.-M.); (L.S.-V.); (G.B.-M.); (D.Q.G.); (A.C.-C.)
| |
Collapse
|
3
|
Buccigrossi V, Poeta M, Cioffi V, Terranova S, Nunziata F, Lo Vecchio A, Guarino A. Lacticaseibacillus rhamnosus GG Counteracts Rotavirus-Induced Ion Secretion and Enterocyte Damage by Inhibiting Oxidative Stress and Apoptosis Through Specific Effects of Living and Postbiotic Preparations. Front Cell Infect Microbiol 2022; 12:854989. [PMID: 35425719 PMCID: PMC9001969 DOI: 10.3389/fcimb.2022.854989] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 03/07/2022] [Indexed: 12/17/2022] Open
Abstract
Background Administration of Lacticaseibacillus rhamnosus GG (LGG) to children with gastroenteritis is recommended by universal guidelines. Rotavirus (RV) causes diarrhea through combined cytotoxic and enterotoxic effects. Aim of this study was to evaluate the mechanisms of efficacy of LGG in an in-vitro model of RV diarrhea in its viable form (LGG) and conditioned medium (mLGG). Methods Ion secretion corresponding to the NSP4 enterotoxic effect, was evaluated by short circuit current (Isc) and the cytotoxic effect by transepithelial electrical resistance (TEER) in Ussing chambers, upon exposure to RV in Caco-2 enterocyte monolayers treated or not with living probiotic or its culture supernatant. Mechanisms of enterotoxic and cytotoxic damage were evaluated including oxidative stress measured by reactive oxygen species, apoptosis evaluated by DAPI and nuclear staining, NFkβ immunofluorescence. Results RV induced Isc increase and TEER decrease, respectively indicating ion secretion and epithelial damage, the two established pathways of diarrhea. Both probiotic preparations reduced both diarrheal effects, but their potency was different. Live LGG was equally effective on both enterotoxic and cytotoxic effect whereas mLGG was highly effective on ion secretion and showed minimal protective effects on cytoskeleton, apoptosis and NFkβ. Conclusions LGG counteracts RV-induced diarrhea by inhibiting both cytotoxic and enterotoxic pathogenic mechanisms. Namely, LGG inhibits chloride secretion by specific moieties secreted in the medium with a direct pharmacologic-like action. This is considered a postbiotic effect. Subsequently, live bacteria exert a probiotic effect protecting the enterocyte structure.
Collapse
|
4
|
Effect of the EM Bokashi® Multimicrobial Probiotic Preparation on the Non-specific Immune Response in Pigs. Probiotics Antimicrob Proteins 2020; 11:1264-1277. [PMID: 30187429 PMCID: PMC6853859 DOI: 10.1007/s12602-018-9460-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The aim of the study was to determine the effect of EM Bokashi® on the phagocytic activity of monocytes and granulocytes, oxidative burst, SWC3, and CD11b + CD18+ expression on monocytes and granulocytes, and the serum concentration of cytokine and lysozyme in pig. 60 Sixty female piglets were divided into two groups: I – control and II – experimental. For the experimental group, a probiotic in the form of the preparation EM Bokashi® was added to the basal feed. Flow cytometry was used to determine selected non-specific immune response parameters, intracellular production of hydrogen peroxide by peripheral granulocytes and monocytes, and surface particles in peripheral blood. The EM Bokashi® preparation used in the study was found to increase phagocytic activity mainly in monocytes, with an increased percentage of phagocytic cells in the experimental group. The highest serum lysozyme concentration in the piglets in the experimental group (2.89 mg/dl), was noted on day 42 of the study. In the group of pigs receiving EM Bokashi®, the percentage of phagocytic cells with SWC3 (monocyte/granulocyte) expression was statistically significantly higher than in the control. The increase in the number of cells with SWC3 (monocyte/granulocyte) expression in the peripheral circulation in combination with the greater capacity of the cells for phagocytosis and respiratory burst confirms that the non-specific immune response was modulated in the pigs supplemented with EM Bokashi®.
Collapse
|
5
|
Effect of Multi-Microbial Probiotic Formulation Bokashi on Pro- and Anti-Inflammatory Cytokines Profile in the Serum, Colostrum and Milk of Sows, and in a Culture of Polymorphonuclear Cells Isolated from Colostrum. Probiotics Antimicrob Proteins 2019; 11:220-232. [PMID: 29305686 PMCID: PMC6449489 DOI: 10.1007/s12602-017-9380-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The use of probiotics in sows during pregnancy and lactation and their impact on the quality of colostrum and milk, as well as the health conditions of their offspring during the rearing period, are currently gaining the attention of researchers. The aim of the study was to determine the effect of Bokashi formulation on the concentrations of pro- and anti-inflammatory cytokines in the serum of sows during pregnancy, in their colostrum and milk, and in a culture of Con-A-stimulated polymorphonuclear cells (PMNs) isolated from the colostrum. The study was conducted on 60 sows aged 2–4 years. EM Bokashi were added to the sows’ feed. The material for the study consisted of peripheral blood, colostrum, and milk. Blood samples were collected from the sows on days 60 and 114 of gestation. Colostrum and milk samples were collected from all sows at 0, 24, 48, 72, 96, 120, 144, and 168 h after parturition. The results indicate that the use of Bokashi as feed additives resulted in increased concentrations of pro-inflammatory cytokines TNF-α and IL-6, which increase the protective capacity of the colostrum by stimulating cellular immune mechanisms protecting the sow and neonates against infection. At the same time, the increased concentrations of cytokines IL-4, IL-10, TGF-β, and of immunoglobulins in the colostrum and milk from sows in the experimental group demonstrate the immunoregulatory effect of Bokashi on Th2 cells and may lead to increased expression of regulatory T cells and polarization of the immune response from Th1 to Th2.
Collapse
|
6
|
Peng X, Wang R, Hu L, Zhou Q, Liu Y, Yang M, Fang Z, Lin Y, Xu S, Feng B, Li J, Jiang X, Zhuo Y, Li H, Wu D, Che L. Enterococcus faecium NCIMB 10415 administration improves the intestinal health and immunity in neonatal piglets infected by enterotoxigenic Escherichia coli K88. J Anim Sci Biotechnol 2019; 10:72. [PMID: 31452881 PMCID: PMC6702752 DOI: 10.1186/s40104-019-0376-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 07/09/2019] [Indexed: 12/11/2022] Open
Abstract
Background This study aimed to investigate the effects of oral administration of Enterococcus faecium NCIMB 10415 (E. faecium) on intestinal development, immunological parameters and gut microbiota of neonatal piglets challenged with enterotoxigenic Escherichia coli K88 (ETEC). A total of 96 1-day-old sow-reared piglets were randomly assigned to 2 groups, with 48 piglets in each group. The piglets were from 16 litters (6 piglets each litter), and 3 piglets each litter were allocated to the E. faecium-supplemented (PRO) group, while the other 3 piglets were allocated to the control (CON) group. After colostrum intake, piglets in the PRO group were orally administrated with 3 × 109 CFU E. faecium per day for a period of one week. On day 8, one piglet per litter from each group was challenged (CON+ETEC, PRO+ETEC) or not (CON-ETEC, PRO-ETEC) with ETEC in a 2 × 2 factorial arrangement of treatments. On day 10 (2 days after challenge), blood and tissue samples were obtained from piglets. Results Before ETEC challenge, there were no significant differences for the average daily gain (ADG) and fecal score between the two groups of piglets. After ETEC challenge, the challenged piglets had greater fecal score compared to the non-challenged piglets, whereas E. faecium administration was able to decrease the fecal score. Piglets challenged with ETEC had shorter villous height, deeper crypt depth, and reduced number of goblet cells in the jejunum and decreased mRNA abundance of claudin-1 in the ileum, whereas increased the percentage of lymphocytes, concentrations of IL-1β in the plasma and TNF-α in the ileal mucosa, as well as increased the mRNA abundances of innate immunity-related genes in the ileum tissue. These deleterious effects caused by ETEC were partly alleviated by feeding E. faecium. In addition, piglets in PRO-ETEC group had decreased the percentage of CD8+ T cells of the peripheral blood when compared to those in CON-ETEC group. Moreover, E. faecium administration increased Verrucomicrobia at phylum level and decreased Bilophila at genus level. Conclusions These results suggest that oral administration of E. faecium alleviated the intestinal injury and diarrhea severity of neonatal piglets challenged by ETEC, partly through improving the intestinal microbiota and immune response. This offers a potential strategy of dietary intervention against intestinal impairment by ETEC in neonatal piglets. Electronic supplementary material The online version of this article (10.1186/s40104-019-0376-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xie Peng
- 1Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan 611130 People's Republic of China
| | - Ru Wang
- 1Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan 611130 People's Republic of China
| | - Liang Hu
- 1Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan 611130 People's Republic of China
| | - Qiang Zhou
- 1Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan 611130 People's Republic of China
| | - Yang Liu
- 1Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan 611130 People's Republic of China
| | - Min Yang
- 1Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan 611130 People's Republic of China.,Animal Husbandry and Veterinary Department, Chengdu Agricultural College, Chengdu, Sichuan 611130 People's Republic of China
| | - Zhengfeng Fang
- 1Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan 611130 People's Republic of China
| | - Yan Lin
- 1Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan 611130 People's Republic of China
| | - Shengyu Xu
- 1Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan 611130 People's Republic of China
| | - Bin Feng
- 1Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan 611130 People's Republic of China
| | - Jian Li
- 1Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan 611130 People's Republic of China
| | - Xuemei Jiang
- 1Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan 611130 People's Republic of China
| | - Yong Zhuo
- 1Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan 611130 People's Republic of China
| | - Hua Li
- 1Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan 611130 People's Republic of China
| | - De Wu
- 1Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan 611130 People's Republic of China
| | - Lianqiang Che
- 1Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan 611130 People's Republic of China
| |
Collapse
|
7
|
Effects of Early Intervention with Maternal Fecal Microbiota and Antibiotics on the Gut Microbiota and Metabolite Profiles of Piglets. Metabolites 2018; 8:metabo8040089. [PMID: 30563199 PMCID: PMC6316024 DOI: 10.3390/metabo8040089] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 11/29/2018] [Accepted: 12/02/2018] [Indexed: 12/24/2022] Open
Abstract
We investigated the effects of early intervention with maternal fecal microbiota and antibiotics on gut microbiota and the metabolites. Five litters of healthy neonatal piglets (Duroc × Landrace × Yorkshire, nine piglets in each litter) were used. Piglets in each litter were orally treated with saline (CO), amoxicillin treatment (AM), or maternal fecal microbiota transplantation (MFMT) on days 1–6, with three piglets in each treatment. Results were compared to the CO group. MFMT decreased the relative abundances of Clostridium sensu stricto and Parabacteroides in the colon on day 7, whereas the abundance of Blautia increased, and the abundance of Corynebacterium in the stomach reduced on day 21. AM reduced the abundance of Arcanobacterium in the stomach on day 7 and reduced the abundances of Streptococcus and Lachnoclostridium in the ileum and colon on day 21, respectively. The metabolite profile indicated that MFMT markedly influenced carbohydrate metabolism and amino acid (AA) metabolism on day 7. On day 21, carbohydrate metabolism and AA metabolism were affected by AM. The results suggest that MFMT and AM discriminatively modulate gastrointestinal microflora and alter the colonic metabolic profiles of piglets and show different effects in the long-term. MFMT showed a location-specific influence on the gastrointestinal microbiota.
Collapse
|
8
|
Early intervention with faecal microbiota transplantation: an effective means to improve growth performance and the intestinal development of suckling piglets. Animal 2018; 13:533-541. [PMID: 29983136 DOI: 10.1017/s1751731118001611] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Recent studies indicate that early postnatal period is a critical window for gut microbiota manipulation to optimise the immunity and body growth. This study investigated the effects of maternal faecal microbiota orally administered to neonatal piglets after birth on growth performance, selected microbial populations, intestinal permeability and the development of intestinal mucosal immune system. In total, 12 litters of crossbred newborn piglets were selected in this study. Litter size was standardised to 10 piglets. On day 1, 10 piglets in each litter were randomly allotted to the faecal microbiota transplantation (FMT) and control groups. Piglets in the FMT group were orally administrated with 2ml faecal suspension of their nursing sow per day from the age of 1 to 3 days; piglets in the control group were treated with the same dose of a placebo (0.1M potassium phosphate buffer containing 10% glycerol (vol/vol)) inoculant. The experiment lasted 21 days. On days 7, 14 and 21, plasma and faecal samples were collected for the analysis of growth-related hormones and cytokines in plasma and lipocalin-2, secretory immunoglobulin A (sIgA), selected microbiota and short-chain fatty acids (SCFAs) in faeces. Faecal microbiota transplantation increased the average daily gain of piglets during week 3 and the whole experiment period. Compared with the control group, the FMT group had increased concentrations of plasma growth hormone and IGF-1 on days 14 and 21. Faecal microbiota transplantation also reduced the incidence of diarrhoea during weeks 1 and 3 and plasma concentrations of zonulin, endotoxin and diamine oxidase activities in piglets on days 7 and 14. The populations of Lactobacillus spp. and Faecalibacterium prausnitzii and the concentrations of faecal and plasma acetate, butyrate and total SCFAs in FMT group were higher than those in the control group on day 21. Moreover, the FMT piglets have higher concentrations of plasma transforming growth factor-β and immunoglobulin G, and faecal sIgA than the control piglets on day 21. These findings indicate that early intervention with maternal faecal microbiota improves growth performance, decreases intestinal permeability, stimulates sIgA secretion, and modulates gut microbiota composition and metabolism in suckling piglets.
Collapse
|
9
|
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
|
10
|
Xie Y, Zhang C, Wang L, Shang Q, Zhang G, Yang W. Effects of dietary supplementation of Enterococcus faecium on growth performance, intestinal morphology, and selected microbial populations of piglets. Livest Sci 2018. [DOI: 10.1016/j.livsci.2018.02.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
11
|
Effects of Early Intervention with Sodium Butyrate on Gut Microbiota and the Expression of Inflammatory Cytokines in Neonatal Piglets. PLoS One 2016; 11:e0162461. [PMID: 27611998 PMCID: PMC5017769 DOI: 10.1371/journal.pone.0162461] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 08/23/2016] [Indexed: 11/19/2022] Open
Abstract
Butyrate in the gut of animals has potential properties including regulating the innate immune, modulating the lipid metabolism, and protecting gut healthy. So far, only limited information on the impact of butyrate on the neonatal is available. This study aimed to investigate effects of oral administration of sodium butyrate (SB) on gut microbiota and the expression of inflammatory cytokine in neonatal piglets. Ten litters of crossbred newborn piglets were randomly allocated to the SB and control (CO) groups, each group consisted of five litters (replicates). Piglets in the SB group were orally administrated with 7 to 13 ml sodium butyrate solution (150 mmol/l) per day from the age of 1 to 7 days, respectively; piglets in the CO group were treated with the same dose of physiological saline. On days 8 and 21 (of age), gut digesta and tissues were collected for the analysis of microbiota, butyrate concentration and gene expression of inflammatory cytokine. Results showed that there was no difference in the butyrate concentration in the gut of piglets on days 8 and 21 between two groups. Real-time PCR assay showed that SB had no effect on the numbers of total bacteria in the stomach, ileum, and colon. MiSeq sequencing of the V3-V4 region of the 16S rRNA gene revealed that SB increased the richness in the stomach and colon, and the diversity of colonic microbiota on day 8 (P < 0.05). Genera Acinetobacter, Actinobacillus, Facklamia, Globicatella, Kocuria, Rothia, unclassified Leptotrichiaceae, unclassified Neisseriaceae, and unclassified Prevotellaceae in the stomach were increased in relative abundance by SB treatment, whereas the abundances of Lactobacillus decreased on day 8 (P < 0.05). At the genus and operational taxonomic unit (OTU) levels, SB had low impact on bacterial community in the ileum and colon on days 8 and 21. SB treatment decreased the expression of IL-6, IL-8, IFN-γ, IL-10, TGF-β, and histone deacetylase 1 (HDAC1) in the ileum of piglets on day 8 (P < 0.05). SB treatment down-regulated the expression of IL-8, IFN-γ, and IL-1β on day 21 (P < 0.05). Correlation analysis on the combined datasets revealed some potential relationships between gut microbiota and the expression of inflammatory cytokines. The results show that early intervention with sodium butyrate can modulate the ileum inflammatory cytokine in neonatal piglets with low impact on intestinal microbial structure, which suggests oral administration of SB may have a benefit role in the health of neonatal piglets.
Collapse
|
12
|
Lactobacillus casei Exerts Anti-Proliferative Effects Accompanied by Apoptotic Cell Death and Up-Regulation of TRAIL in Colon Carcinoma Cells. PLoS One 2016; 11:e0147960. [PMID: 26849051 PMCID: PMC4744000 DOI: 10.1371/journal.pone.0147960] [Citation(s) in RCA: 198] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Accepted: 01/10/2016] [Indexed: 12/11/2022] Open
Abstract
Probiotic microorganisms such as lactic acid bacteria (LAB) exert a number of strain-specific health-promoting activities attributed to their immunomodulatory, anti-inflammatory and anti-carcinogenic properties. Despite recent attention, our understanding of the biological processes involved in the beneficial effects of LAB strains is still limited. To this end, the present study investigated the growth-inhibitory effects of Lactobacillus casei ATCC 393 against experimental colon cancer. Administration of live Lactobacillus casei (as well as bacterial components thereof) on murine (CT26) and human (HT29) colon carcinoma cell lines raised a significant concentration- and time-dependent anti-proliferative effect, determined by cell viability assays. Specifically, a dramatic decrease in viability of colon cancer cells co-incubated with 10(9) CFU/mL L. casei for 24 hours was detected (78% for HT29 and 52% for CT26 cells). In addition, live L. casei induced apoptotic cell death in both cell lines as revealed by annexin V and propidium iodide staining. The significance of the in vitro anti-proliferative effects was further confirmed in an experimental tumor model. Oral daily administration of 10(9) CFU live L. casei for 13 days significantly inhibited in vivo growth of colon carcinoma cells, resulting in approximately 80% reduction in tumor volume of treated mice. Tumor growth inhibition was accompanied by L. casei-driven up-regulation of the TNF-related apoptosis-inducing ligand TRAIL and down-regulation of Survivin. Taken together, these findings provide evidence for beneficial tumor-inhibitory, anti-proliferative and pro-apoptotic effects driven by this probiotic LAB strain.
Collapse
|
13
|
Barfod KK, Vrankx K, Mirsepasi-Lauridsen HC, Hansen JS, Hougaard KS, Larsen ST, Ouwenhand AC, Krogfelt KA. The Murine Lung Microbiome Changes During Lung Inflammation and Intranasal Vancomycin Treatment. Open Microbiol J 2015; 9:167-79. [PMID: 26668669 PMCID: PMC4676059 DOI: 10.2174/1874285801509010167] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Revised: 09/01/2015] [Accepted: 09/02/2015] [Indexed: 12/15/2022] Open
Abstract
Most microbiome research related to airway diseases has focused on the gut microbiome. This is despite advances
in culture independent microbial identification techniques revealing that even healthy lungs possess a unique dynamic
microbiome. This conceptual change raises the question; if lung diseases could be causally linked to local dysbiosis
of the local lung microbiota. Here, we manipulate the murine lung and gut microbiome, in order to show that the lung microbiota
can be changed experimentally. We have used four different approaches: lung inflammation by exposure to carbon
nano-tube particles, oral probiotics and oral or intranasal exposure to the antibiotic vancomycin. Bacterial DNA was
extracted from broncho-alveolar and nasal lavage fluids, caecum samples and compared by DGGE. Our results show that:
the lung microbiota is sex dependent and not just a reflection of the gut microbiota, and that induced inflammation can
change lung microbiota. This change is not transferred to offspring. Oral probiotics in adult mice do not change lung microbiome
detectible by DGGE. Nasal vancomycin can change the lung microbiome preferentially, while oral exposure
does not. These observations should be considered in future studies of the causal relationship between lung microbiota
and lung diseases.
Collapse
Affiliation(s)
| | - Katleen Vrankx
- Applied Maths, Keistraat 120, 9830 Sint-Martens-Latem, Belgium
| | | | - Jitka Stilund Hansen
- National Research Centre for the Working Environment, Lersø parkallé 105, 2100 Denmark
| | - Karin Sørig Hougaard
- National Research Centre for the Working Environment, Lersø parkallé 105, 2100 Denmark
| | - Søren Thor Larsen
- National Research Centre for the Working Environment, Lersø parkallé 105, 2100 Denmark
| | - Arthur C Ouwenhand
- Active Nutrition, Dupont Nutrition & Health, Sokeritehtaantie 20, 02460 Kantvik Finland
| | | |
Collapse
|
14
|
Hou C, Liu H, Zhang J, Zhang S, Yang F, Zeng X, Thacker PA, Zhang G, Qiao S. Intestinal microbiota succession and immunomodulatory consequences after introduction of Lactobacillus reuteri I5007 in neonatal piglets. PLoS One 2015; 10:e0119505. [PMID: 25775260 PMCID: PMC4361599 DOI: 10.1371/journal.pone.0119505] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Accepted: 01/14/2015] [Indexed: 01/06/2023] Open
Abstract
Seventy-two, suckling piglets, obtained from 9 litters standardized to 8 piglets, were assigned to 1 of 3 treatments (n = 24) to compare short-term, early administration with intermittent, longer-term administration of Lactobacillus reuteri I5007. The treatments were a control (given a placebo of 0.1% peptone water from day 1 to 5) or treatments in which 1.7 × 1010 CFU L. reuteri was administrated either daily for 4 days starting on day 1 or every 4th day from day 1 to 17. Five piglets per treatment were killed at 3 time points (day 7, 14 and 21). Denaturing Gradient Electrophoresis of ileal digesta revealed an increase in the presence of L. reuteri I5007 and Clostridium lentocellum (on day 14 and 21) in the every 4th-day treatment and Actinobacillus porcinus (on day 7 and 14) in both L. reuteri treatments, while reducing the abundance of E. coli on day 21 in the every 4th-day treatment. Real-time qPCR of ileal digesta showed an increase in Bifidobacterium spp. on day 14 for both L. reuteri I5007 treatments. An increase in the concentration of lactic acid and a lower pH was observed in the first 4-day treatment on day 7 and the every 4th day treatment on day 14. The relative abundance of mRNA for TGF-β was increased while that for IFN-γ was decreased in the mesenteric lymph nodes of piglets treated with L. reuteri every 4th day. In conclusion, early intervention with L. reuteri increases the presence of beneficial bacteria and decreases the presence of undesirable microbes in the lower gastrointestinal tract. The changes appear to be mediated by altering the intestinal pH through lactic acid production resulting in favorable bacterial species colonization. A prolonged duration of treatment (i.e. every 4th day) would appear to be superior to treatment only during the first 4 days.
Collapse
Affiliation(s)
- Chengli Hou
- State Key Laboratory of Animal Nutrition, China Agricultural University, No. 2, Yuanmingyuan West Road, Beijing 100193, China
| | - Hong Liu
- State Key Laboratory of Animal Nutrition, China Agricultural University, No. 2, Yuanmingyuan West Road, Beijing 100193, China
| | - Jiang Zhang
- State Key Laboratory of Animal Nutrition, China Agricultural University, No. 2, Yuanmingyuan West Road, Beijing 100193, China
| | - Shihai Zhang
- State Key Laboratory of Animal Nutrition, China Agricultural University, No. 2, Yuanmingyuan West Road, Beijing 100193, China
| | - Fengjuan Yang
- State Key Laboratory of Animal Nutrition, China Agricultural University, No. 2, Yuanmingyuan West Road, Beijing 100193, China
| | - Xiangfang Zeng
- State Key Laboratory of Animal Nutrition, China Agricultural University, No. 2, Yuanmingyuan West Road, Beijing 100193, China
| | - Philip A Thacker
- Department of Animal and Poultry Science, University of Saskatchewan, Saskatoon, Canada
| | - Guolong Zhang
- Department of Animal Science, Oklahoma State University, Stillwater, Oklahoma, United States of America
| | - Shiyan Qiao
- State Key Laboratory of Animal Nutrition, China Agricultural University, No. 2, Yuanmingyuan West Road, Beijing 100193, China
- * E-mail:
| |
Collapse
|
15
|
Yang F, Hou C, Zeng X, Qiao S. The use of lactic Acid bacteria as a probiotic in Swine diets. Pathogens 2015; 4:34-45. [PMID: 25633489 PMCID: PMC4384071 DOI: 10.3390/pathogens4010034] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Revised: 12/22/2014] [Accepted: 01/22/2015] [Indexed: 02/07/2023] Open
Abstract
As the resistance of pathogens to antibiotics and the possibility of antibiotic residues in animal products attract increasing attention, the interest in the use of alternatives to in-feed antibiotics has been growing. Recent research with Lactic acid bacteria (LAB) in pigs suggests that LAB provide a potential alternative to antibiotic strategies. LAB include Lactobacillus species, Bifidobacterium spp, Bacillus spp, and some other microbes. LAB can adjust the intestinal environment, inhibit or kill pathogens in the gastrointestinal tract and improve the microbial balance in the intestine, as well as regulate intestinal mucosal immunity and maintain intestinal barrier function, thereby benefiting the health of pigs. The related mechanisms for these effects of LAB may include producing microbicidal substances with effects against gastrointestinal pathogens and other harmful microbes, competing with pathogens for binding sites on the intestinal epithelial cell surface and mucin as well as stimulating the immune system. In this review, the characteristics of LAB and their probiotic effects in newborn piglets, weaned piglets, growing pigs and sows are documented.
Collapse
Affiliation(s)
- Fengjuan Yang
- State Key Laboratory of Animal Nutrition, China Agricultural University, No. 2, Yuanmingyuan West Road, Beijing 100193, China.
| | - Chengli Hou
- State Key Laboratory of Animal Nutrition, China Agricultural University, No. 2, Yuanmingyuan West Road, Beijing 100193, China.
| | - Xiangfang Zeng
- State Key Laboratory of Animal Nutrition, China Agricultural University, No. 2, Yuanmingyuan West Road, Beijing 100193, China.
| | - Shiyan Qiao
- State Key Laboratory of Animal Nutrition, China Agricultural University, No. 2, Yuanmingyuan West Road, Beijing 100193, China.
| |
Collapse
|
16
|
Liu H, Zhang J, Zhang S, Yang F, Thacker PA, Zhang G, Qiao S, Ma X. Oral administration of Lactobacillus fermentum I5007 favors intestinal development and alters the intestinal microbiota in formula-fed piglets. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2014; 62:860-6. [PMID: 24404892 DOI: 10.1021/jf403288r] [Citation(s) in RCA: 132] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
The present study was conducted to evaluate the effects of early administration of Lactobacillus fermentum I5007 on intestinal development and microbial composition in the gastrointestinal tract using a neonatal piglet model. Full-term 4 day old piglets, fed with milk replacer, were divided into a control group (given placebo of 0.1% peptone water) and a L. fermentum I5007 group (dosed daily with 6 × 10(9) CFU/mL L. fermentum I5007). The experiment lasted 14 days. On day 14, a significant increase in the jejunum villous height (583 ± 33 vs 526 ± 18) and increases in the concentrations of butyrate (7.55 ± 0.55 vs 5.33 ± 0.39) and branched chain fatty acids in the colonic digesta were observed in piglets in the L. fermentum I5007 treatment (P < 0.05). mRNA expression of IL-1β (1.29 ± 0.29 vs. 0.62 ± 0.07) in the ileum were lower after 14 days of treatment with L. fermentum I5007. Denaturing gradient gel electrophoresis (DGGE) revealed that L. fermentum I5007 affected the colonic microbial communities on day 14 and, in particular, reduced numbers of Clostridium sp. L. fermentum I5007 play a positive role in gut development in neonatal piglets by modulating microbial composition, intestinal development, and immune status. L. fermentum I5007 may be useful as a probiotic for application in neonatal piglets.
Collapse
Affiliation(s)
- Hong Liu
- State Key Laboratory of Animal Nutrition, China Agricultural University , No. 2, Yuanmingyuan West Road, Beijing 100193, China
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Brasili E, Mengheri E, Tomassini A, Capuani G, Roselli M, Finamore A, Sciubba F, Marini F, Miccheli A. Lactobacillus acidophilus La5 and Bifidobacterium lactis Bb12 induce different age-related metabolic profiles revealed by 1H-NMR spectroscopy in urine and feces of mice. J Nutr 2013; 143:1549-57. [PMID: 23946343 DOI: 10.3945/jn.113.177105] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Age-related dysbioses of intestinal microbiota and decline in the overall metabolic homeostasis are frequently found in the elderly. Probiotic supplementation may represent a way to prevent or reduce the senescence-associated metabolic disorders. The present study evaluated the metabolic impact of Lactobacillus acidophilus La5 and Bifidobacterium lactis Bb12 supplementation in relation to age by analyzing urine and feces metabolic profiles using (1)H-nuclear magnetic resonance spectroscopy and multivariate analysis. Adult (3 mo old) and aged (16 mo old) mice received an oral supplementation of the 2 probiotics (1 × 10(9) colony-forming units/d each) or phosphate buffered saline (control) daily for 30 d. Urine and feces were collected for 48 h before the end of the study. Partial least squares-discriminant analysis showed that the urinary discriminant metabolites for the probiotic treatment included higher dimethylglycine in adult and aged mice, lower sarcosine and nicotinate in adult mice, higher N-methylnicotinamide in adult mice and lower N-methylnicotinamide in aged mice compared with their controls. These results indicate a probiotic-induced modulation of homocysteine and NAD metabolism pathways, which have important implications because these pathways are involved in essential cellular processes that can be altered in senescence. The probiotic supplementation also modified the fecal metabolic profiles, inducing in both adult and aged mice higher 4-hydroxyphenylacetate and lower xylose in treated mice compared with their control mice, whereas valerate was greater in treated adult mice and lower in treated aged mice compared with their controls. The ANOVA simultaneous component analysis on urinary and fecal metabolic profiling showed an age × treatment interaction (P < 0.05), confirming the age-related modulation of the metabolic response to probiotic supplementation. The results suggest that L. acidophilus and B. lactis may prevent or reduce age-related metabolic dysfunction.
Collapse
Affiliation(s)
- Elisa Brasili
- National Research Institute on Food and Nutrition (INRAN), Department of Nutritional Sciences, Rome, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Gosai V, Ambalam P, Raman M, Kothari CR, Kothari RK, Vyas BRM, Sheth NR. Protective effect of Lactobacillus rhamnosus 231 against N-Methyl-N'-nitro-N-nitrosoguanidine in animal model. Gut Microbes 2013; 2:319-25. [PMID: 22157237 DOI: 10.4161/gmic.18755] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
The protective effect of Lactobacillus rhamnosus 231 (Lr 231) against potent carcinogen N-Methyl-N'-Nitro-N-Nitrosoguanidine (MNNG) in the rat model is studied. Daily feeding with Lr 231 improved the body weight of male Wistar rats compared with control groups. Fecal azoreductase (p < 0.001) and nitroreductase (p < 0.01) enzyme activity decreased significantly in Lr 231 group in comparison with control groups that received only phosphate buffer or MNNG. Oral administration of MNNG led to a significant increase in Glutathione transferase (GST) while Glutathione reductase (GSH) showed decreased activity. Conversely, feeding Lr 231 showed significantly increased GSH and decreased GST activity in comparison to the MNNG group, emphasizing the protection provided by Lr 231 against MNNG. Histopathological analysis of liver, spleen and colon showed decreased signs of inflammation in the Lr 231 group. The present study highlights that inclusion of active Lr 231 in regular diets could be used to prevent MNNG induced colon carcinoma.
Collapse
Affiliation(s)
- Viral Gosai
- Department of Pharmacy, Saurashtra University, Rajkot, India
| | | | | | | | | | | | | |
Collapse
|
19
|
Nagalingam NA, Lynch SV. Role of the microbiota in inflammatory bowel diseases. Inflamm Bowel Dis 2012; 18:968-84. [PMID: 21936031 DOI: 10.1002/ibd.21866] [Citation(s) in RCA: 213] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2011] [Accepted: 07/26/2011] [Indexed: 12/16/2022]
Abstract
Studying the role of the human microbiome as it relates to human health status has revolutionized our view of microbial community contributions to a large number of diseases, particularly chronic inflammatory disorders. The lower gastrointestinal (GI) tract houses trillions of microbial cells representing a large diversity of species in relatively well-defined phylogenetic ratios that are associated with maintenance of key aspects of host physiology and immune homeostasis. It is not surprising, therefore, that many GI inflammatory diseases, including inflammatory bowel disease (IBD), are associated with substantial changes in the composition of these microbial assemblages, either as a cause or consequence of host inflammatory response. Here we review current knowledge in the emerging field of human microbiome research as it relates to IBD, specifically focusing on Crohn's disease (CD) and ulcerative colitis (UC). We discuss bacteriotherapeutic efforts to restore GI microbial assemblage integrity via probiotic supplementation of IBD patients, and speculate on future directions for the field.
Collapse
Affiliation(s)
- Nabeetha A Nagalingam
- Colitis and Crohn's Disease Microbiome Research Core, Division of Gastroenterology, University of California, San Francisco, Calfornia 94143-0538, USA
| | | |
Collapse
|
20
|
Williams BL, Hornig M, Buie T, Bauman ML, Cho Paik M, Wick I, Bennett A, Jabado O, Hirschberg DL, Lipkin WI. Impaired carbohydrate digestion and transport and mucosal dysbiosis in the intestines of children with autism and gastrointestinal disturbances. PLoS One 2011; 6:e24585. [PMID: 21949732 PMCID: PMC3174969 DOI: 10.1371/journal.pone.0024585] [Citation(s) in RCA: 350] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2011] [Accepted: 08/14/2011] [Indexed: 12/11/2022] Open
Abstract
Gastrointestinal disturbances are commonly reported in children with autism, complicate clinical management, and may contribute to behavioral impairment. Reports of deficiencies in disaccharidase enzymatic activity and of beneficial responses to probiotic and dietary therapies led us to survey gene expression and the mucoepithelial microbiota in intestinal biopsies from children with autism and gastrointestinal disease and children with gastrointestinal disease alone. Ileal transcripts encoding disaccharidases and hexose transporters were deficient in children with autism, indicating impairment of the primary pathway for carbohydrate digestion and transport in enterocytes. Deficient expression of these enzymes and transporters was associated with expression of the intestinal transcription factor, CDX2. Metagenomic analysis of intestinal bacteria revealed compositional dysbiosis manifest as decreases in Bacteroidetes, increases in the ratio of Firmicutes to Bacteroidetes, and increases in Betaproteobacteria. Expression levels of disaccharidases and transporters were associated with the abundance of affected bacterial phylotypes. These results indicate a relationship between human intestinal gene expression and bacterial community structure and may provide insights into the pathophysiology of gastrointestinal disturbances in children with autism.
Collapse
Affiliation(s)
- Brent L Williams
- Center for Infection and Immunity, Columbia University, New York, New York, United States of America
| | | | | | | | | | | | | | | | | | | |
Collapse
|